PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 7778849-6 1995 Robust 32-kd bands corresponding to heme oxygenase-1 were observed by Western blotting of protein extracts derived from AD temporal cortex and hippocampus after sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Sodium Dodecyl Sulfate 161-183 heme oxygenase 1 Homo sapiens 36-52 7778849-6 1995 Robust 32-kd bands corresponding to heme oxygenase-1 were observed by Western blotting of protein extracts derived from AD temporal cortex and hippocampus after sodium dodecyl sulfate-polyacrylamide gel electrophoresis. polyacrylamide 184-198 heme oxygenase 1 Homo sapiens 36-52 8163576-7 1994 Heavy metal ions and thiol reagents were also highly potent inducers of HO-1 in human RPE cells. Metals 6-11 heme oxygenase 1 Homo sapiens 72-76 7857260-0 1995 Differential control of the metal-mediated activation of the human heme oxygenase-1 and metallothionein IIA genes. Metals 28-33 heme oxygenase 1 Homo sapiens 67-83 7857260-1 1995 Heme oxygenase-1, an essential enzyme in heme catabolism, and metallothionein IIA, a small metal-binding protein with clusters of cysteins, are remarkably induced in HeLa cells following the treatment with cadmium or zinc. Cadmium 206-213 heme oxygenase 1 Homo sapiens 0-16 7857260-3 1995 Here we showed by transient expression assays that the cadmium-responsive element (CdRE) of the human heme oxygenase-1 gene is not responsive to zinc, whereas a metal-regulatory element (MRE) of the human metallothionein IIA gene is able to respond to either cadmium or zinc. Cadmium 55-62 heme oxygenase 1 Homo sapiens 102-118 7857260-3 1995 Here we showed by transient expression assays that the cadmium-responsive element (CdRE) of the human heme oxygenase-1 gene is not responsive to zinc, whereas a metal-regulatory element (MRE) of the human metallothionein IIA gene is able to respond to either cadmium or zinc. cdre 83-87 heme oxygenase 1 Homo sapiens 102-118 7857260-3 1995 Here we showed by transient expression assays that the cadmium-responsive element (CdRE) of the human heme oxygenase-1 gene is not responsive to zinc, whereas a metal-regulatory element (MRE) of the human metallothionein IIA gene is able to respond to either cadmium or zinc. Metals 161-166 heme oxygenase 1 Homo sapiens 102-118 7857260-3 1995 Here we showed by transient expression assays that the cadmium-responsive element (CdRE) of the human heme oxygenase-1 gene is not responsive to zinc, whereas a metal-regulatory element (MRE) of the human metallothionein IIA gene is able to respond to either cadmium or zinc. Cadmium 259-266 heme oxygenase 1 Homo sapiens 102-118 8030754-1 1994 Heme oxygenase-1 is an important enzyme that degrades heme, a pro-oxidant, leading to the formation of antioxidant molecules. Heme 54-58 heme oxygenase 1 Homo sapiens 0-16 8016102-3 1994 Heme treatment increased both HO activity and HO-1 mRNA in the human erythroleukemic cell line K562. Heme 0-4 heme oxygenase 1 Homo sapiens 46-50 7703255-3 1995 The purified, truncated hHO-1/heme complex is spectroscopically indistinguishable from that of the rat enzyme and converts heme to biliverdin when reconstituted with rat liver cytochrome P450 reductase. Heme 30-34 heme oxygenase 1 Homo sapiens 24-29 7703255-3 1995 The purified, truncated hHO-1/heme complex is spectroscopically indistinguishable from that of the rat enzyme and converts heme to biliverdin when reconstituted with rat liver cytochrome P450 reductase. Heme 123-127 heme oxygenase 1 Homo sapiens 24-29 7703255-3 1995 The purified, truncated hHO-1/heme complex is spectroscopically indistinguishable from that of the rat enzyme and converts heme to biliverdin when reconstituted with rat liver cytochrome P450 reductase. Biliverdine 131-141 heme oxygenase 1 Homo sapiens 24-29 7525927-0 1994 Induction of heart heme oxygenase-1 (HSP32) by hyperthermia: possible role in stress-mediated elevation of cyclic 3":5"-guanosine monophosphate. Cyclic GMP 107-143 heme oxygenase 1 Homo sapiens 19-35 7525927-0 1994 Induction of heart heme oxygenase-1 (HSP32) by hyperthermia: possible role in stress-mediated elevation of cyclic 3":5"-guanosine monophosphate. Cyclic GMP 107-143 heme oxygenase 1 Homo sapiens 37-42 7525927-2 1994 Heme oxygenase (HO) isozymes, HO-1 (HSP32) and HO-2, catalyze the rate limiting step in the only known pathway in eukaryotes for the generation of the potential cellular message, carbon monoxide, and the antioxidant, bilirubin. Carbon Monoxide 179-194 heme oxygenase 1 Homo sapiens 30-34 7525927-2 1994 Heme oxygenase (HO) isozymes, HO-1 (HSP32) and HO-2, catalyze the rate limiting step in the only known pathway in eukaryotes for the generation of the potential cellular message, carbon monoxide, and the antioxidant, bilirubin. Carbon Monoxide 179-194 heme oxygenase 1 Homo sapiens 36-41 7525927-2 1994 Heme oxygenase (HO) isozymes, HO-1 (HSP32) and HO-2, catalyze the rate limiting step in the only known pathway in eukaryotes for the generation of the potential cellular message, carbon monoxide, and the antioxidant, bilirubin. Bilirubin 217-226 heme oxygenase 1 Homo sapiens 30-34 7525927-2 1994 Heme oxygenase (HO) isozymes, HO-1 (HSP32) and HO-2, catalyze the rate limiting step in the only known pathway in eukaryotes for the generation of the potential cellular message, carbon monoxide, and the antioxidant, bilirubin. Bilirubin 217-226 heme oxygenase 1 Homo sapiens 36-41 8163576-7 1994 Heavy metal ions and thiol reagents were also highly potent inducers of HO-1 in human RPE cells. Sulfhydryl Compounds 21-26 heme oxygenase 1 Homo sapiens 72-76 8455037-1 1993 In mammalian systems, the heme oxygenase (HO) isozymes HO-1 (HSP32) and HO-2 oxidatively cleave the heme molecule to produce bile pigments and carbon monoxide. Heme 26-30 heme oxygenase 1 Homo sapiens 61-66 19912946-0 1993 Heme Oxygenase, a Likely Regulator of cGMP Production in the Brain: Induction in Vivo of HO-1 Compensates for Depression in NO Synthase Activity. Cyclic GMP 38-42 heme oxygenase 1 Homo sapiens 89-93 19912946-2 1993 The sole source of CO in mammalian systems is the alpha-meso carbon bridge of the heme molecule cleaved by heme oxygenase isozymes, HO-1 and HO-2. Carbon 61-67 heme oxygenase 1 Homo sapiens 132-145 19912946-2 1993 The sole source of CO in mammalian systems is the alpha-meso carbon bridge of the heme molecule cleaved by heme oxygenase isozymes, HO-1 and HO-2. Heme 82-86 heme oxygenase 1 Homo sapiens 132-145 19912946-10 1993 Increased capability of select cerebellar cell populations to generate CO, as indicated by an increase in their HO-1 protein content, points to the active role of this isozyme in maintenance of cGMP level under stress conditions, when nitric oxide production is compromised. Carbon Monoxide 71-73 heme oxygenase 1 Homo sapiens 112-116 8146161-1 1994 Oxidative stress of human skin fibroblasts by treatment with ultraviolet A (UVA) radiation has been shown to lead to an increase in levels of the heme catabolizing enzyme heme oxygenase 1 [heme, hydrogen-donor:oxygen oxidoreductase (alpha-methene-oxidizing, hydroxylating), EC 1.14.99.3] and the iron storage protein ferritin. Heme 146-150 heme oxygenase 1 Homo sapiens 171-187 8146161-1 1994 Oxidative stress of human skin fibroblasts by treatment with ultraviolet A (UVA) radiation has been shown to lead to an increase in levels of the heme catabolizing enzyme heme oxygenase 1 [heme, hydrogen-donor:oxygen oxidoreductase (alpha-methene-oxidizing, hydroxylating), EC 1.14.99.3] and the iron storage protein ferritin. Hydrogen 195-203 heme oxygenase 1 Homo sapiens 171-187 8146161-3 1994 Pretreating cells with heme oxygenase 1 antisense oligonucleotide inhibited the irradiation-dependent induction of both the heme oxygenase I enzyme and ferritin and abolished the protective effect of preirradiation. Oligonucleotides 50-65 heme oxygenase 1 Homo sapiens 23-39 8455037-1 1993 In mammalian systems, the heme oxygenase (HO) isozymes HO-1 (HSP32) and HO-2 oxidatively cleave the heme molecule to produce bile pigments and carbon monoxide. Carbon Monoxide 143-158 heme oxygenase 1 Homo sapiens 61-66 1754738-2 1991 We observed an increase in mRNA levels of heat-shock protein (hsp) 70, hsp 90, hsp 32 and metallothionein after treatment of GOTO cells with cadmium, although the time courses of the changes of individual mRNA of the heat-shock proteins and metallothionein were somewhat different from each other. Cadmium 141-148 heme oxygenase 1 Homo sapiens 79-85 33818898-6 2021 Deoxynivalenol (IC50 = 20 muM) activated mitochondrial apoptotic pathway by modulating antioxidant protein expressions (Nrf2, HO-1 and NQO1). deoxynivalenol 0-14 heme oxygenase 1 Homo sapiens 126-130 33775716-0 2021 The signaling pathway PI3K/Akt/Nrf2/HO-1 plays a role in the mitochondrial protection promoted by astaxanthin in the SH-SY5Y cells exposed to hydrogen peroxide. astaxanthine 98-109 heme oxygenase 1 Homo sapiens 36-40 33775716-0 2021 The signaling pathway PI3K/Akt/Nrf2/HO-1 plays a role in the mitochondrial protection promoted by astaxanthin in the SH-SY5Y cells exposed to hydrogen peroxide. Hydrogen Peroxide 142-159 heme oxygenase 1 Homo sapiens 36-40 33775716-9 2021 Inhibition of the PI3K/Akt or of the HO-1 enzyme abolished the AST-induced mitochondrial protection in cells challenged with H2O2. astaxanthine 63-66 heme oxygenase 1 Homo sapiens 37-41 33775716-9 2021 Inhibition of the PI3K/Akt or of the HO-1 enzyme abolished the AST-induced mitochondrial protection in cells challenged with H2O2. Hydrogen Peroxide 125-129 heme oxygenase 1 Homo sapiens 37-41 33775716-11 2021 Thus, we suggest that AST promotes mitochondrial protection by a mechanism dependent on the PI3K/Akt/Nrf2/HO-1 signaling pathway in SH-SY5Y cells exposed to H2O2. Hydrogen Peroxide 157-161 heme oxygenase 1 Homo sapiens 106-110 2052613-1 1991 Catalytic activity of heme oxygenase (heme, hydrogen-donor:oxygen oxidoreductase, EC 1.14.99.3) isozymes, HO-1 and HO-2, permits production of physiologic isomers of bile pigments. Hydrogen 44-52 heme oxygenase 1 Homo sapiens 106-119 33761605-0 2021 Heme oxygenase-1 inducer hemin does not inhibit SARS-CoV-2 virus infection. Hemin 25-30 heme oxygenase 1 Homo sapiens 0-16 33761605-3 2021 Given that hemin is a natural inducer of the HO-1 gene, the aim of this study was to develop an in vitro assay to analyze the antiviral potency of hemin against SARS-CoV-2 infection. Hemin 11-16 heme oxygenase 1 Homo sapiens 45-49 33761605-3 2021 Given that hemin is a natural inducer of the HO-1 gene, the aim of this study was to develop an in vitro assay to analyze the antiviral potency of hemin against SARS-CoV-2 infection. Hemin 147-152 heme oxygenase 1 Homo sapiens 45-49 33761605-7 2021 Hemin administration to the culture medium induced a high induction in the expression of the HO-1 gene that was stronger in Vero-E6 macaque-derived cells than in the human Calu-3 cell line. Hemin 0-5 heme oxygenase 1 Homo sapiens 93-97 33761605-9 2021 In conclusion, although exposure to hemin induced strong HO-1 up-regulation, this effect was unable to inhibit or delay the progression of SARS-CoV-2 infection in two epithelial cell lines susceptible to infection. Hemin 36-41 heme oxygenase 1 Homo sapiens 57-61 33788639-6 2021 ECs pre-treated with different concentrations RA and co-cultured with thapsigargin-induced ER stressed pancreatic beta-cells showed increased levels of Nrf2 and its downstream targets such as heme oxygenase-1 (HO-1) and NADPH-quinone oxidoreductase-1 (NQO-1), and reduction of ER stress evinced by the decreased levels of glucose-regulated protein (GRP) 78 and C/ERB homologous protein (CHOP). Thapsigargin 70-82 heme oxygenase 1 Homo sapiens 192-208 33818552-9 2021 The protective effect of chrysin on sepsis-induced cardiac dysfunction was blocked by ZnPP, which is a HO-1 blocker. chrysin 25-32 heme oxygenase 1 Homo sapiens 103-107 33818552-9 2021 The protective effect of chrysin on sepsis-induced cardiac dysfunction was blocked by ZnPP, which is a HO-1 blocker. zinc protoporphyrin 86-90 heme oxygenase 1 Homo sapiens 103-107 33788639-6 2021 ECs pre-treated with different concentrations RA and co-cultured with thapsigargin-induced ER stressed pancreatic beta-cells showed increased levels of Nrf2 and its downstream targets such as heme oxygenase-1 (HO-1) and NADPH-quinone oxidoreductase-1 (NQO-1), and reduction of ER stress evinced by the decreased levels of glucose-regulated protein (GRP) 78 and C/ERB homologous protein (CHOP). Thapsigargin 70-82 heme oxygenase 1 Homo sapiens 210-214 33805292-4 2021 We systematically reviewed published evidence on HMOX1 polymorphisms in perinatal diseases and clarified their possible significant contribution to neonatal jaundice development, presumably due to their direct effect of inducing HO enzymatic activity in the bilirubin-producing pathway. Bilirubin 258-267 heme oxygenase 1 Homo sapiens 49-54 33807391-6 2021 IL-8 mRNA expression was analyzed in saliva-stimulated THP-1 cells treated with CAPE and the heme oxygenase-1 (HO-1) inhibitor tin-protoporphyrin (SnPP). tin protoporphyrin IX 127-145 heme oxygenase 1 Homo sapiens 93-109 33807391-6 2021 IL-8 mRNA expression was analyzed in saliva-stimulated THP-1 cells treated with CAPE and the heme oxygenase-1 (HO-1) inhibitor tin-protoporphyrin (SnPP). tin protoporphyrin IX 127-145 heme oxygenase 1 Homo sapiens 111-115 33807391-11 2021 CAPE induced HO-1 expression and inhibited IKK2, IkappaBalpha, and NF-kappaB phosphorylation. caffeic acid phenethyl ester 0-4 heme oxygenase 1 Homo sapiens 13-17 33807391-12 2021 Blocking HO-1 decreased the anti-inflammatory activity of CAPE. caffeic acid phenethyl ester 58-62 heme oxygenase 1 Homo sapiens 9-13 33808635-7 2021 SiRNA-mediated knockdown of HO-1 significantly increased the oxidative stress induced by palmitate, whereas pre-treatment with SnCl2, a well-known HO-1 inducer, significantly decreased it. Palmitates 89-98 heme oxygenase 1 Homo sapiens 28-32 33774859-5 2021 HO-1, Trx-1 and Nrf-2 expression levels significantly increased on pre-treatment with SS-31 compared with the H2 O2 group. arginyl-2,'6'-dimethyltyrosyl-lysyl-phenylalaninamide 86-91 heme oxygenase 1 Homo sapiens 0-4 33770188-11 2021 We demonstrated that PSF enhanced Nrf2 activation and heme oxygenase-1 (HO-1) expression via extracellular signal-regulated kinase (ERK) and Akt signaling in HRMECs, which subsequently resulted in intracellular ROS scavenging. Reactive Oxygen Species 211-214 heme oxygenase 1 Homo sapiens 72-76 33815094-0 2021 Elevated Heme Oxygenase-1 Correlates With Increased Brain Iron Deposition Measured by Quantitative Susceptibility Mapping and Decreased Hemoglobin in Patients With Parkinson"s Disease. Iron 58-62 heme oxygenase 1 Homo sapiens 9-25 33815094-3 2021 Objective: To explore the association of the level of HO-1 with brain iron deposition and low level of HGB in PD. Iron 70-74 heme oxygenase 1 Homo sapiens 54-58 33808635-8 2021 Moreover, SB203580, a selective p38 inhibitor, reduced HO-1 mRNA expression and increased palmitate-induced oxidative stress in HepG2 cells. SB 203580 10-18 heme oxygenase 1 Homo sapiens 55-59 33815094-11 2021 There was a significantly positive correlation between the serum HO-1 concentration and iron deposition within SN, an inverse correlation between the serum HO-1 concentration and HGB level in PD patients. Iron 88-92 heme oxygenase 1 Homo sapiens 65-69 33808635-9 2021 These results suggest that the HO-1-mediated anti-oxidative stress compensatory reaction plays an essential role against saturated fatty acid-induced lipotoxicity in the liver. Fatty Acids 121-141 heme oxygenase 1 Homo sapiens 31-35 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Heme 64-68 heme oxygenase 1 Homo sapiens 0-16 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Heme 64-68 heme oxygenase 1 Homo sapiens 18-22 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Biliverdine 101-111 heme oxygenase 1 Homo sapiens 0-16 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Biliverdine 101-111 heme oxygenase 1 Homo sapiens 18-22 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 0-16 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 18-22 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Iron 139-143 heme oxygenase 1 Homo sapiens 0-16 33804125-1 2021 Heme oxygenase-1 (HO-1) plays a vital role in the catabolism of heme and yields equimolar amounts of biliverdin, carbon monoxide, and free iron. Iron 139-143 heme oxygenase 1 Homo sapiens 18-22 33803317-5 2021 Heme oxygenase-1 (HO-1) is an iron-dependent cytoprotective enzyme that functions as the inducible form of HO. Iron 30-34 heme oxygenase 1 Homo sapiens 0-16 33803317-5 2021 Heme oxygenase-1 (HO-1) is an iron-dependent cytoprotective enzyme that functions as the inducible form of HO. Iron 30-34 heme oxygenase 1 Homo sapiens 18-22 33803289-7 2021 The DMF treatment was associated with no changes in virus replication; higher expressions of the antioxidant enzymes NQO1, GPX1, and HO-1 in the brain and PRDX1 and HO-2 in the spleen; lower levels of 8-OHdG and 3NT; a lower optical redox ratio. Dimethyl Fumarate 4-7 heme oxygenase 1 Homo sapiens 133-137 33803317-6 2021 HO-1 and its metabolites carbon monoxide and biliverdin contribute towards the maintenance of redox homeostasis. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-4 33803317-6 2021 HO-1 and its metabolites carbon monoxide and biliverdin contribute towards the maintenance of redox homeostasis. Biliverdine 45-55 heme oxygenase 1 Homo sapiens 0-4 33801315-8 2021 Moreover, we showed that onopordopicrin promoted the expression of the nuclear factor-erythroid-2-related factor 2 (Nrf2) downstream target protein heme oxygenase-1 (HO-1) in muscle cells. onopordopicrin 25-39 heme oxygenase 1 Homo sapiens 148-164 33801315-8 2021 Moreover, we showed that onopordopicrin promoted the expression of the nuclear factor-erythroid-2-related factor 2 (Nrf2) downstream target protein heme oxygenase-1 (HO-1) in muscle cells. onopordopicrin 25-39 heme oxygenase 1 Homo sapiens 166-170 33801315-9 2021 By using siRNA, we demonstrated that the inhibition of the expression of Nrf2 reduced the protective effect of onopordopicrin, indicating that the activation of the Nrf2/HO-1 signaling pathway mediates the antioxidant effect of onopordopicrin in primary human muscle cells. onopordopicrin 111-125 heme oxygenase 1 Homo sapiens 170-174 33801315-9 2021 By using siRNA, we demonstrated that the inhibition of the expression of Nrf2 reduced the protective effect of onopordopicrin, indicating that the activation of the Nrf2/HO-1 signaling pathway mediates the antioxidant effect of onopordopicrin in primary human muscle cells. onopordopicrin 228-242 heme oxygenase 1 Homo sapiens 170-174 33801351-4 2021 In this study, we found that 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), one of the final products of COX-2, induced upregulation of vascular endothelial growth factor (VEGF) and capillary formation and migration through nuclear factor erythroid 2-related factor 2 (NRF2)-dependent heme oxygenase-1 (HO-1) induction in MCF-7 cells. 14-prostaglandin j2 46-65 heme oxygenase 1 Homo sapiens 287-303 33801351-4 2021 In this study, we found that 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), one of the final products of COX-2, induced upregulation of vascular endothelial growth factor (VEGF) and capillary formation and migration through nuclear factor erythroid 2-related factor 2 (NRF2)-dependent heme oxygenase-1 (HO-1) induction in MCF-7 cells. 14-prostaglandin j2 46-65 heme oxygenase 1 Homo sapiens 305-309 33801351-4 2021 In this study, we found that 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), one of the final products of COX-2, induced upregulation of vascular endothelial growth factor (VEGF) and capillary formation and migration through nuclear factor erythroid 2-related factor 2 (NRF2)-dependent heme oxygenase-1 (HO-1) induction in MCF-7 cells. 15-deoxy-delta(12,14)-prostaglandin J2 67-75 heme oxygenase 1 Homo sapiens 287-303 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Heme 79-83 heme oxygenase 1 Homo sapiens 0-16 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Heme 79-83 heme oxygenase 1 Homo sapiens 18-22 33801351-4 2021 In this study, we found that 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), one of the final products of COX-2, induced upregulation of vascular endothelial growth factor (VEGF) and capillary formation and migration through nuclear factor erythroid 2-related factor 2 (NRF2)-dependent heme oxygenase-1 (HO-1) induction in MCF-7 cells. 15-deoxy-delta(12,14)-prostaglandin J2 67-75 heme oxygenase 1 Homo sapiens 305-309 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Heme 79-83 heme oxygenase 1 Homo sapiens 35-40 33801351-7 2021 In conclusion, the elevated levels of 15d-PGJ2 during inflammatory response activate VEGF expression through NRF2-driven induction of HO-1 in human breast cancer cells, proposing a novel mechanism underlying the oncogenic function of 15d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 38-46 heme oxygenase 1 Homo sapiens 134-138 33801351-7 2021 In conclusion, the elevated levels of 15d-PGJ2 during inflammatory response activate VEGF expression through NRF2-driven induction of HO-1 in human breast cancer cells, proposing a novel mechanism underlying the oncogenic function of 15d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 234-242 heme oxygenase 1 Homo sapiens 134-138 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 0-16 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Carbon Monoxide 135-150 heme oxygenase 1 Homo sapiens 15-31 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 18-22 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 35-40 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. ammonium ferrous sulfate 93-97 heme oxygenase 1 Homo sapiens 0-16 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. ammonium ferrous sulfate 93-97 heme oxygenase 1 Homo sapiens 18-22 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. ammonium ferrous sulfate 93-97 heme oxygenase 1 Homo sapiens 35-40 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Biliverdine 103-113 heme oxygenase 1 Homo sapiens 0-16 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Biliverdine 103-113 heme oxygenase 1 Homo sapiens 18-22 33804563-1 2021 Heme oxygenase-1 (HO-1, encoded by HMOX1) is a cytoprotective enzyme degrading heme into CO, Fe2+, and biliverdin. Biliverdine 103-113 heme oxygenase 1 Homo sapiens 35-40 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Heme 15-19 heme oxygenase 1 Homo sapiens 33-37 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Carbon Monoxide 135-150 heme oxygenase 1 Homo sapiens 33-37 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Biliverdine 152-162 heme oxygenase 1 Homo sapiens 15-31 33589574-5 2021 Our results showed that high expression of HO-1 and CD163 were detected in TAMs for 57.9% (73/126) and 61.9% (78/126) of the studied patients, and both of them were significantly associated with worse survival. tams 75-79 heme oxygenase 1 Homo sapiens 43-47 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Biliverdine 152-162 heme oxygenase 1 Homo sapiens 33-37 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Bilirubin 163-172 heme oxygenase 1 Homo sapiens 15-31 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Bilirubin 163-172 heme oxygenase 1 Homo sapiens 33-37 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Iron 183-187 heme oxygenase 1 Homo sapiens 15-31 33587911-4 2021 In the future, heme oxygenase-1 (HO-1) may also become a candidate ILD biomarker; it is a 32-kDa heat shock protein converting heme to carbon monoxide, biliverdin/bilirubin, and free iron to play a role in the pulmonary cytoprotective reaction in response to various stimuli. Iron 183-187 heme oxygenase 1 Homo sapiens 33-37 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Heme 77-81 heme oxygenase 1 Homo sapiens 0-22 33232417-6 2020 Moreover, IOT also enhanced the expression of the GCLC, GCLM, HO-1, NQO1 and Trx-1 proteins, which mostly depends on the nuclear translation of Nrf2 and reduced expression of the Keap1 protein. homoorientin 10-13 heme oxygenase 1 Homo sapiens 62-66 29662820-7 2018 Interestingly, the increased heme oxygenase 1 expression under hypoxic conditions was decreased by selenium treatment, whereas superoxide dismutase expression was increased in trophoblast cells by selenium treatment for 72 hours, regardless of hypoxia. Selenium 99-107 heme oxygenase 1 Homo sapiens 29-45 33763175-7 2021 Furthermore, farrerol markedly activated Nrf2, thereby increasing the levels of antioxidant enzymes downstream of Nrf2, such as HO-1, NQO1, and GCLM. farrerol 13-21 heme oxygenase 1 Homo sapiens 128-132 33763175-8 2021 Knockdown of Nrf2 with a specific siRNA successfully suppressed farrerol-mediated HO-1 transcription and partially abolished the cytoprotective effect on ARPE-19 cells. farrerol 64-72 heme oxygenase 1 Homo sapiens 82-86 31624724-14 2019 We observed that genes encoding C/EBP delta, FOS-like antigen 1, c-Jun, and heme oxygenase-1 (HO-1) were most highly induced in the HCT116 cells following 15d-PGJ2 treatment. 15-deoxy-delta(12,14)-prostaglandin J2 155-163 heme oxygenase 1 Homo sapiens 76-92 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Heme 77-81 heme oxygenase 1 Homo sapiens 35-40 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Carbon Monoxide 87-102 heme oxygenase 1 Homo sapiens 0-22 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Carbon Monoxide 87-102 heme oxygenase 1 Homo sapiens 35-40 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. ammonium ferrous sulfate 123-127 heme oxygenase 1 Homo sapiens 0-22 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. ammonium ferrous sulfate 123-127 heme oxygenase 1 Homo sapiens 35-40 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Biliverdine 133-143 heme oxygenase 1 Homo sapiens 0-22 29628790-1 2018 Heme oxygenase-1 (HO-1, encoded by HMOX1) through degradation of pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin, exhibits cytoprotective, anti-apoptotic and anti-inflammatory properties. Biliverdine 133-143 heme oxygenase 1 Homo sapiens 35-40 29628790-6 2018 Metalloporphyrins are widely used in in vitro studies, however, they are unselective and may exert serious side effects including an increase in HMOX1 mRNA level. Metalloporphyrins 0-17 heme oxygenase 1 Homo sapiens 145-150 25303561-5 2015 The "braking system" includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-gamma-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. Carbon Monoxide 180-195 heme oxygenase 1 Homo sapiens 92-97 26331632-6 2015 Moreover, hydrogenation of the double bond of the functional alpha,beta-unsaturated carbonyl group of AVAs eliminated their effects on HO-1 expression, suggesting that this group is crucial for the antioxidant activity of AVAs. avenanthramide-2C 102-106 heme oxygenase 1 Homo sapiens 135-139 26331632-6 2015 Moreover, hydrogenation of the double bond of the functional alpha,beta-unsaturated carbonyl group of AVAs eliminated their effects on HO-1 expression, suggesting that this group is crucial for the antioxidant activity of AVAs. avenanthramide-2C 222-226 heme oxygenase 1 Homo sapiens 135-139 26629073-9 2015 Moreover, when HO-1 was interfered with siRNA, the effects of RLM on the levels of ROS and MMP were diminished, indicating that HO-1 induction was necessary for the function of RLM. Reactive Oxygen Species 83-86 heme oxygenase 1 Homo sapiens 15-19 26629073-9 2015 Moreover, when HO-1 was interfered with siRNA, the effects of RLM on the levels of ROS and MMP were diminished, indicating that HO-1 induction was necessary for the function of RLM. Reactive Oxygen Species 83-86 heme oxygenase 1 Homo sapiens 128-132 26629073-12 2015 RLM inhibits ROS production and elevates MMP, through the induction of HO-1 expression, in LECs under high glucose condition. Glucose 107-114 heme oxygenase 1 Homo sapiens 71-75 25493608-10 2015 This could reflect decreases in cellular heme caused by the massive induction by arsenite of heme oxygenase mRNA (HMOX1; 68-fold increase), the rate-limiting enzyme in heme catabolism. Heme 41-45 heme oxygenase 1 Homo sapiens 114-119 25493608-10 2015 This could reflect decreases in cellular heme caused by the massive induction by arsenite of heme oxygenase mRNA (HMOX1; 68-fold increase), the rate-limiting enzyme in heme catabolism. arsenite 81-89 heme oxygenase 1 Homo sapiens 114-119 26331632-0 2015 Oat avenanthramides induce heme oxygenase-1 expression via Nrf2-mediated signaling in HK-2 cells. avenanthramide-2C 4-19 heme oxygenase 1 Homo sapiens 27-43 26331632-2 2015 Here, we investigated whether AVAs affect heme oxygenase-1 (HO-1) expression through the activation of Nrf2 translocation. avenanthramide-2C 30-34 heme oxygenase 1 Homo sapiens 42-58 26331632-2 2015 Here, we investigated whether AVAs affect heme oxygenase-1 (HO-1) expression through the activation of Nrf2 translocation. avenanthramide-2C 30-34 heme oxygenase 1 Homo sapiens 60-64 26331632-4 2015 Furthermore, we found that AVA-induced HO-1 expression is mediated by Nrf2 translocation. avenanthramide-2C 27-30 heme oxygenase 1 Homo sapiens 39-43 26331632-5 2015 The addition of N-acetylcysteine (NAC), but not specific inhibitors of p38 (SB202190), PI3K (LY294002), and MEK1 (PD098059) attenuated AVA-induced HO-1 expression, demonstrating an important role for reactive oxygen species, but not PI3K or MAPK activation, in activating the HO-1 pathway. Acetylcysteine 16-32 heme oxygenase 1 Homo sapiens 147-151 26331632-5 2015 The addition of N-acetylcysteine (NAC), but not specific inhibitors of p38 (SB202190), PI3K (LY294002), and MEK1 (PD098059) attenuated AVA-induced HO-1 expression, demonstrating an important role for reactive oxygen species, but not PI3K or MAPK activation, in activating the HO-1 pathway. Acetylcysteine 16-32 heme oxygenase 1 Homo sapiens 276-280 25303561-5 2015 The "braking system" includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-gamma-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. Carbon Monoxide 197-199 heme oxygenase 1 Homo sapiens 92-97 25303561-5 2015 The "braking system" includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-gamma-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. Hydrogen Sulfide 205-222 heme oxygenase 1 Homo sapiens 92-97 25303561-5 2015 The "braking system" includes the protective pathways of haem oxygenase 1 (also referred as Hmox1 or HO-1) and cystathionine-gamma-lyase (also known as CSE or Cth), which generate carbon monoxide (CO) and hydrogen sulphide (H2S) respectively. Hydrogen Sulfide 224-227 heme oxygenase 1 Homo sapiens 92-97 19161347-1 2009 HO-1 (haem oxygenase 1) is an essential antioxidant enzyme in the cell that exerts its effects through removal of pro-oxidant haem groups and the formation of antioxidant molecules and carbon monoxide. Carbon Monoxide 185-200 heme oxygenase 1 Homo sapiens 0-22 19161347-2 2009 The electrophilic cyclopentenone 15d-PGJ2 (15-deoxy-Delta(12,14)-prostaglandin J2) induces the expression of HO-1 protein through the covalent modification of protein thiols. cyclopentenone 15d-pgj2 18-41 heme oxygenase 1 Homo sapiens 109-113 19161347-7 2009 IBTP had no effect on basal HO-1 levels, but effectively blocked HO-1 induction by a variety of reagents including haemin, iodoacetamide and 15d-PGJ2. Iodoacetamide 123-136 heme oxygenase 1 Homo sapiens 65-69 19161347-7 2009 IBTP had no effect on basal HO-1 levels, but effectively blocked HO-1 induction by a variety of reagents including haemin, iodoacetamide and 15d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 141-149 heme oxygenase 1 Homo sapiens 65-69 19161347-2 2009 The electrophilic cyclopentenone 15d-PGJ2 (15-deoxy-Delta(12,14)-prostaglandin J2) induces the expression of HO-1 protein through the covalent modification of protein thiols. 15-deoxy-delta(12,14)-prostaglandin J2 43-81 heme oxygenase 1 Homo sapiens 109-113 19161347-9 2009 However, IBTP prevented the 15d-PGJ2-dependent increases in HO-1 mRNA and protein. ibtp 9-13 heme oxygenase 1 Homo sapiens 60-64 19161347-12 2009 In addition, IBTP significantly enhanced the toxicity of high concentrations of 15d-PGJ2, suggesting that loss of mitochondrial control of HO-1 leads to increased susceptibility to electrophilic stress in endothelial cells. ibtp 13-17 heme oxygenase 1 Homo sapiens 139-143 19161347-2 2009 The electrophilic cyclopentenone 15d-PGJ2 (15-deoxy-Delta(12,14)-prostaglandin J2) induces the expression of HO-1 protein through the covalent modification of protein thiols. Sulfhydryl Compounds 167-173 heme oxygenase 1 Homo sapiens 109-113 19161347-12 2009 In addition, IBTP significantly enhanced the toxicity of high concentrations of 15d-PGJ2, suggesting that loss of mitochondrial control of HO-1 leads to increased susceptibility to electrophilic stress in endothelial cells. 15-deoxy-delta(12,14)-prostaglandin J2 80-88 heme oxygenase 1 Homo sapiens 139-143 19161347-3 2009 It has been shown that specific thiol residues of the redox-sensor Keap1 (Kelch-like ECH-associated protein 1) are modified by 15d-PGJ2, leading to activation of the transcription factor Nrf-2 (nuclear factor-erythroid 2 p45 subunit-related factor 2) and up-regulation of genes under control of the electrophile-response element, including HO-1. Sulfhydryl Compounds 32-37 heme oxygenase 1 Homo sapiens 340-344 19161347-6 2009 In order to determine the role of mitochondrial protein thiol modification in HO-1 induction, we used the mitochondrial-targeted thiol-reactive compound IBTP [(4-iodobutyl)triphenylphosphonium]. ibtp 153-157 heme oxygenase 1 Homo sapiens 78-82 19161347-7 2009 IBTP had no effect on basal HO-1 levels, but effectively blocked HO-1 induction by a variety of reagents including haemin, iodoacetamide and 15d-PGJ2. ibtp 0-4 heme oxygenase 1 Homo sapiens 65-69 19161347-7 2009 IBTP had no effect on basal HO-1 levels, but effectively blocked HO-1 induction by a variety of reagents including haemin, iodoacetamide and 15d-PGJ2. Hemin 115-121 heme oxygenase 1 Homo sapiens 65-69 34919942-0 2022 Catalpol alleviates myocardial ischemia reperfusion injury by activating the Nrf2/HO-1 signaling pathway. catalpol 0-8 heme oxygenase 1 Homo sapiens 82-86 12709575-4 2003 The research was developed, switching off inducible nitric oxide synthase (iNOS) expression through antisense oligonucleotide transfection by studying the possible coregulation in the expression of HSP32 (also named HO-1), HSP70, and iNOS and their involvement in the induction of DNA damage. Oligonucleotides 110-125 heme oxygenase 1 Homo sapiens 198-203 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. tin protoporphyrin IX 10-17 heme oxygenase 1 Homo sapiens 106-122 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. tin protoporphyrin IX 10-17 heme oxygenase 1 Homo sapiens 124-128 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. cdt 80-83 heme oxygenase 1 Homo sapiens 106-122 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. cdt 80-83 heme oxygenase 1 Homo sapiens 124-128 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. Heme 139-143 heme oxygenase 1 Homo sapiens 106-122 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. Heme 139-143 heme oxygenase 1 Homo sapiens 124-128 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. Bilirubin 174-183 heme oxygenase 1 Homo sapiens 106-122 34863556-7 2022 Moreover, TinPPIX further augments the free heme level along with amplifies the CDT efficacy by disabling heme oxygenase-1 (HO-1)-mediated heme conversion into antioxidative bilirubin. Bilirubin 174-183 heme oxygenase 1 Homo sapiens 124-128 19497345-8 2009 Interestingly, palmatine increased heme oxygenase (HO)-1 induction in human aortic endothelial cells. palmatine 15-24 heme oxygenase 1 Homo sapiens 35-56 34920080-10 2022 Quercetin, an HO-1 inducer, reduced syncytia formation and spike protein expression. Quercetin 0-9 heme oxygenase 1 Homo sapiens 14-18 34919942-11 2022 CONCLUSION: In summary, the findings suggest that Catalpol exerted significant cardioprotective effects following myocardial ischemia, possibly through the activation of the Nrf2/HO-1 signaling pathway. catalpol 50-58 heme oxygenase 1 Homo sapiens 179-183 34953799-0 2022 Overexpressed mitogen-and stress-activated protein kinase 1 promotes the resistance of cytarabine in acute myeloid leukemia through brahma related gene 1-mediated upregulation of heme oxygenase-1. Cytarabine 87-97 heme oxygenase 1 Homo sapiens 179-195 34614244-8 2022 Suppression of high glucose-induced cell proliferation by lutein was not associated with apoptosis induction, but it was linked with inhibition of hyperglycemia-mediated elevated ROS and upregulated expression of high glucose-mediated repressed heme oxygenase 1 (HO1). Glucose 218-225 heme oxygenase 1 Homo sapiens 245-261 34953799-10 2022 However, the increased cell sensitivity induced by MSK1 downregulation was reversed by the induction of HO-1 inducer Hemin. Hemin 117-122 heme oxygenase 1 Homo sapiens 104-108 34614244-8 2022 Suppression of high glucose-induced cell proliferation by lutein was not associated with apoptosis induction, but it was linked with inhibition of hyperglycemia-mediated elevated ROS and upregulated expression of high glucose-mediated repressed heme oxygenase 1 (HO1). Glucose 218-225 heme oxygenase 1 Homo sapiens 263-266 34614244-8 2022 Suppression of high glucose-induced cell proliferation by lutein was not associated with apoptosis induction, but it was linked with inhibition of hyperglycemia-mediated elevated ROS and upregulated expression of high glucose-mediated repressed heme oxygenase 1 (HO1). Glucose 20-27 heme oxygenase 1 Homo sapiens 245-261 34614244-8 2022 Suppression of high glucose-induced cell proliferation by lutein was not associated with apoptosis induction, but it was linked with inhibition of hyperglycemia-mediated elevated ROS and upregulated expression of high glucose-mediated repressed heme oxygenase 1 (HO1). Glucose 20-27 heme oxygenase 1 Homo sapiens 263-266 34129218-0 2022 Astaxanthin Relieves Busulfan-Induced Oxidative Apoptosis in Cultured Human Spermatogonial Stem Cells by Activating the Nrf-2/HO-1 pathway. astaxanthine 0-11 heme oxygenase 1 Homo sapiens 126-130 34613549-7 2022 The results showed that oleuropein significantly decreased levels of the inflammatory mediator cyclooxygenase-2 and increased anti-inflammatory protein HO-1 expression. oleuropein 24-34 heme oxygenase 1 Homo sapiens 152-156 34565648-0 2022 Corrigendum to "Neuroprotective effect of phosphocreatine on oxidative stress and mitochondrial dysfunction induced apoptosis in vitro and in vivo: Involvement of dual PI3K/Akt and Nrf2/HO-1 pathways" (Free Radic. Phosphocreatine 42-57 heme oxygenase 1 Homo sapiens 186-190 34664771-0 2022 Bisphenol A induced apoptosis via oxidative stress generation involved Nrf2/HO-1 pathway and mitochondrial dependent pathways in human retinal pigment epithelium (ARPE-19) cells. bis(4-hydroxyphenyl)sulfone 0-9 heme oxygenase 1 Homo sapiens 76-80 34742568-12 2022 Mechanistically, mitoquinone was found to enhance the activity of Nrf2 and heme oxygenase-1 and to induce nuclear translocation of Nrf2 in human peritoneal mesothelial cells. mitoquinone 17-28 heme oxygenase 1 Homo sapiens 75-91 34813774-5 2022 Interestingly, salidroside partly reversed DHT mediated effects, via stimulation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and the downstream antioxidant proteins heme oxygenase-1(HO-1) and quinine oxidoreductase 1(NQO1). rhodioloside 15-26 heme oxygenase 1 Homo sapiens 210-214 34813774-5 2022 Interestingly, salidroside partly reversed DHT mediated effects, via stimulation of nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and the downstream antioxidant proteins heme oxygenase-1(HO-1) and quinine oxidoreductase 1(NQO1). Dihydrotestosterone 43-46 heme oxygenase 1 Homo sapiens 210-214 34813774-7 2022 Mechanistically, AMP-activated protein kinase (AMPK) was identified to be the upstream signaling involved in salidroside-induced Nrf2 activation, as silencing of AMPK partly prevented the upregulation of Nrf2 and the downstream proteins HO-1 and NQO1. rhodioloside 109-120 heme oxygenase 1 Homo sapiens 237-241 34664771-9 2022 Our results showed that expression of HO-1 and Nrf2 suppressed by BPA. bisphenol A 66-69 heme oxygenase 1 Homo sapiens 38-42 34664771-13 2022 Taken together, these results suggest that BPA induced ARPE-19 cells via oxidative stress, which was associated with down regulated Nrf2/HO-1 pathway, and the mitochondria dependent apoptotic signaling pathway. bisphenol A 43-46 heme oxygenase 1 Homo sapiens 137-141 34848368-12 2022 Probenecid, a well-known UA-lowering drug, significantly suppressed the activation of TXNIP and Nrf2/HO-1 signaling. Probenecid 0-10 heme oxygenase 1 Homo sapiens 101-105 34716926-0 2022 Surfactin reduces particulate matter-induced VCAM-1-dependent monocyte adhesion in human gingival fibroblasts by increasing Nrf2-dependent HO-1 expression. surfactin peptide 0-9 heme oxygenase 1 Homo sapiens 139-143 34716926-3 2022 As surfactin has potential to induce Nrf2 activation and HO-1 expression, this study aimed to investigate the anti-inflammatory effects of surfactin on PM-exposed human gingival fibroblasts (HGFs) and signaling pathways engaged by surfactin. surfactin peptide 3-12 heme oxygenase 1 Homo sapiens 57-61 34716926-3 2022 As surfactin has potential to induce Nrf2 activation and HO-1 expression, this study aimed to investigate the anti-inflammatory effects of surfactin on PM-exposed human gingival fibroblasts (HGFs) and signaling pathways engaged by surfactin. surfactin peptide 139-148 heme oxygenase 1 Homo sapiens 57-61 34716926-3 2022 As surfactin has potential to induce Nrf2 activation and HO-1 expression, this study aimed to investigate the anti-inflammatory effects of surfactin on PM-exposed human gingival fibroblasts (HGFs) and signaling pathways engaged by surfactin. surfactin peptide 231-240 heme oxygenase 1 Homo sapiens 57-61 34906793-7 2022 Our study revealed that m-beta-CoV upregulated Park7, RelA, Nrf2, and Hmox1 transcripts involved in ROS production and antioxidant pathways, describing the possible nexus between oxidative pathways, MMPs, and TIMP in m-beta-CoV-induced neuroinflammation. ros 100-103 heme oxygenase 1 Homo sapiens 70-75 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Heme 76-80 heme oxygenase 1 Homo sapiens 0-16 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Heme 76-80 heme oxygenase 1 Homo sapiens 18-22 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Iron 91-95 heme oxygenase 1 Homo sapiens 0-16 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Iron 91-95 heme oxygenase 1 Homo sapiens 18-22 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 0-16 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 18-22 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Biliverdine 117-127 heme oxygenase 1 Homo sapiens 0-16 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Biliverdine 117-127 heme oxygenase 1 Homo sapiens 18-22 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Bilirubin 166-175 heme oxygenase 1 Homo sapiens 0-16 34841438-1 2022 Heme oxygenase-1 (HO-1) is an inducible cytoprotective enzyme that degrades heme into free iron, carbon monoxide and biliverdin, which is then rapidly converted into bilirubin. Bilirubin 166-175 heme oxygenase 1 Homo sapiens 18-22 34841438-4 2022 In the 5"-non-coding region of the HO-1 gene, there are two polymorphic sites, namely the (GT)n dinucleotide and T(-413)A single nucleotide polymorphism sites, which regulate the transcriptional activity of HO-1. gt)n dinucleotide 91-108 heme oxygenase 1 Homo sapiens 35-39 34841438-4 2022 In the 5"-non-coding region of the HO-1 gene, there are two polymorphic sites, namely the (GT)n dinucleotide and T(-413)A single nucleotide polymorphism sites, which regulate the transcriptional activity of HO-1. gt)n dinucleotide 91-108 heme oxygenase 1 Homo sapiens 207-211 34964128-0 2021 Cyanidin-3-glucoside protects against high glucose-induced injury in human nucleus pulposus cells by regulating the Nrf2/HO-1 signaling. cyanidin-3-o-glucoside 0-20 heme oxygenase 1 Homo sapiens 121-125 34927515-8 2022 Furthermore, 5-ALA is a metabolic precursor of heme, which is a potent inducer of the enzyme heme oxygenase-1, the levels of which are decreased in patients with severe COVID-19. 5-amino levulinic acid 13-18 heme oxygenase 1 Homo sapiens 93-109 34927515-8 2022 Furthermore, 5-ALA is a metabolic precursor of heme, which is a potent inducer of the enzyme heme oxygenase-1, the levels of which are decreased in patients with severe COVID-19. Heme 47-51 heme oxygenase 1 Homo sapiens 93-109 34927515-9 2022 Oral administration of 5-ALA/SFC induced heme oxygenase-1 in the peripheral blood of uninfected healthy individuals. 5-amino levulinic acid 23-28 heme oxygenase 1 Homo sapiens 41-57 34927515-9 2022 Oral administration of 5-ALA/SFC induced heme oxygenase-1 in the peripheral blood of uninfected healthy individuals. SFC 29-32 heme oxygenase 1 Homo sapiens 41-57 34964128-15 2021 Summarily, C3G exerted a protective effect on ROS-mediated cellular damage in HNPCs under HG condition, which was attributed to the induction of the Nrf2/HO-1 signaling pathway. ros 46-49 heme oxygenase 1 Homo sapiens 154-158 34973332-2 2022 Humans express two isoforms of HO: the inducible HO-1, which is up-regulated in response to various stressors, including excess heme, and the constitutive HO-2. Heme 128-132 heme oxygenase 1 Homo sapiens 49-53 34973332-8 2022 When heme is in excess, HO-1 is induced and both HO-2 and HO-1 can provide protection from heme toxicity via enzymatic degradation. Heme 5-9 heme oxygenase 1 Homo sapiens 24-28 34973332-8 2022 When heme is in excess, HO-1 is induced and both HO-2 and HO-1 can provide protection from heme toxicity via enzymatic degradation. Heme 5-9 heme oxygenase 1 Homo sapiens 58-62 34973332-8 2022 When heme is in excess, HO-1 is induced and both HO-2 and HO-1 can provide protection from heme toxicity via enzymatic degradation. Heme 91-95 heme oxygenase 1 Homo sapiens 58-62 34959690-0 2021 From Far West to East: Joining the Molecular Architecture of Imidazole-like Ligands in HO-1 Complexes. imidazole 61-70 heme oxygenase 1 Homo sapiens 87-91 34874275-0 2021 HIF1A-induced heme oxygenase 1 promotes the survival of decidual stromal cells against excess heme-mediated oxidative stress. Heme 94-98 heme oxygenase 1 Homo sapiens 14-30 34874275-1 2021 Heme oxygenase 1 (HO-1, encoded by the HMOX1 gene), is the rate-limiting enzyme that catalyzes heme degradation, and it has been reported to exert antioxidative effects. Heme 95-99 heme oxygenase 1 Homo sapiens 0-16 34874275-1 2021 Heme oxygenase 1 (HO-1, encoded by the HMOX1 gene), is the rate-limiting enzyme that catalyzes heme degradation, and it has been reported to exert antioxidative effects. Heme 95-99 heme oxygenase 1 Homo sapiens 18-22 34874275-1 2021 Heme oxygenase 1 (HO-1, encoded by the HMOX1 gene), is the rate-limiting enzyme that catalyzes heme degradation, and it has been reported to exert antioxidative effects. Heme 95-99 heme oxygenase 1 Homo sapiens 39-44 34874275-6 2021 However, addition of the competitive HO-1 inhibitor ZnPP resulted in an increase in HIF1A expression. zinc protoporphyrin 52-56 heme oxygenase 1 Homo sapiens 37-41 34874275-8 2021 As an HO-1 inducer, cobalt protoporphyrin IX (CoPP) decreased ROS production and significantly reversed the inhibitory effect of H2O2 on cell viability. cobaltiprotoporphyrin 20-44 heme oxygenase 1 Homo sapiens 6-10 34874275-8 2021 As an HO-1 inducer, cobalt protoporphyrin IX (CoPP) decreased ROS production and significantly reversed the inhibitory effect of H2O2 on cell viability. cobaltiprotoporphyrin 46-50 heme oxygenase 1 Homo sapiens 6-10 34874275-8 2021 As an HO-1 inducer, cobalt protoporphyrin IX (CoPP) decreased ROS production and significantly reversed the inhibitory effect of H2O2 on cell viability. Reactive Oxygen Species 62-65 heme oxygenase 1 Homo sapiens 6-10 34874275-8 2021 As an HO-1 inducer, cobalt protoporphyrin IX (CoPP) decreased ROS production and significantly reversed the inhibitory effect of H2O2 on cell viability. Hydrogen Peroxide 129-133 heme oxygenase 1 Homo sapiens 6-10 34874275-10 2021 This study suggests that the upregulation of HO-1 expression via HIF1A protects DSCs against excessive heme-mediated oxidative stress. Heme 103-107 heme oxygenase 1 Homo sapiens 45-49 34496009-6 2021 HO-1 inhibition by tin porphyrin eliminated ex vivo HIV-1 restriction. tin porphyrin 19-32 heme oxygenase 1 Homo sapiens 0-4 34954677-7 2021 In addition, BPA increased the levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) and decreased antioxidant enzymes nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) levels, resulting in oxidative stress. bisphenol A 13-16 heme oxygenase 1 Homo sapiens 191-207 34954677-7 2021 In addition, BPA increased the levels of intracellular reactive oxygen species (ROS) and malondialdehyde (MDA) and decreased antioxidant enzymes nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) levels, resulting in oxidative stress. bisphenol A 13-16 heme oxygenase 1 Homo sapiens 209-213 34928467-3 2021 This study aimed to investigate the predictive value of the total bilirubin level, a marker of heme oxygenase-1 enzyme activity, in determining myocarditis in patients with COVID-19. Bilirubin 66-75 heme oxygenase 1 Homo sapiens 95-111 34946766-0 2021 Nanogold Particles Suppresses 5-Flurouracil-Induced Renal Injury: An Insight into the Modulation of Nrf-2 and Its Downstream Targets, HO-1 and gamma-GCS. 5-flurouracil 30-43 heme oxygenase 1 Homo sapiens 134-138 34912465-0 2021 Shikonin Alleviates Endothelial Cell Injury Induced by ox-LDL via AMPK/Nrf2/HO-1 Signaling Pathway. shikonin 0-8 heme oxygenase 1 Homo sapiens 76-80 34912465-7 2021 Compared to the ox-LDL group, SKN treatment improves cell viability, alleviates cell apoptosis and oxidative stress injury, and upregulates the protein levels of p-AMPK, Nrf2, and HO-1. shikonin 30-33 heme oxygenase 1 Homo sapiens 180-184 34943041-2 2021 The parasite regulates the host immune system by inducing a strong Th2 and regulatory T (Treg) cell immune response through mechanisms that might involve the expression or activity of heme-oxygenase-1 (HO-1), the rate-limiting enzyme in the catabolism of free heme that also has immunoregulatory and antioxidant properties. th2 67-70 heme oxygenase 1 Homo sapiens 202-206 34912465-8 2021 Compound C, si-Nrf2, and si-HO-1 administration inhibits the AMPK/Nrf2/HO-1 signaling pathway, increases ROS generation, and inhibits the antagonistic effect of SKN on ox-LDL-induced HUVECs damage. Silicon 12-14 heme oxygenase 1 Homo sapiens 71-75 34912465-8 2021 Compound C, si-Nrf2, and si-HO-1 administration inhibits the AMPK/Nrf2/HO-1 signaling pathway, increases ROS generation, and inhibits the antagonistic effect of SKN on ox-LDL-induced HUVECs damage. Reactive Oxygen Species 105-108 heme oxygenase 1 Homo sapiens 28-32 34912465-8 2021 Compound C, si-Nrf2, and si-HO-1 administration inhibits the AMPK/Nrf2/HO-1 signaling pathway, increases ROS generation, and inhibits the antagonistic effect of SKN on ox-LDL-induced HUVECs damage. shikonin 161-164 heme oxygenase 1 Homo sapiens 28-32 34866353-14 2022 Heme was the most abundant metabolite in degenerated regions, whereas Heme oxygenase-1 (HO-1), which catabolizes heme, was found in intact regions. Heme 113-117 heme oxygenase 1 Homo sapiens 70-86 34866353-14 2022 Heme was the most abundant metabolite in degenerated regions, whereas Heme oxygenase-1 (HO-1), which catabolizes heme, was found in intact regions. Heme 113-117 heme oxygenase 1 Homo sapiens 88-92 34866353-15 2022 Higher HO-1 levels correlated with lower heme accumulation. Heme 41-45 heme oxygenase 1 Homo sapiens 7-11 34943041-2 2021 The parasite regulates the host immune system by inducing a strong Th2 and regulatory T (Treg) cell immune response through mechanisms that might involve the expression or activity of heme-oxygenase-1 (HO-1), the rate-limiting enzyme in the catabolism of free heme that also has immunoregulatory and antioxidant properties. Heme 260-264 heme oxygenase 1 Homo sapiens 202-206 34634993-1 2021 BACKGROUND: Heme oxygenase (HO)-1 is a rate-limiting enzyme for degrading heme into carbon monoxide. Heme 74-78 heme oxygenase 1 Homo sapiens 12-33 34940692-7 2021 EEB decreased UVB-induced reactive oxygen species (ROS) generation by upregulating glutathione peroxidase 1 (GPx1) and heme oxygenase-1 (HO-1) expression via nuclear factor erythroid-2-related factor 2 (Nrf2) activation in Hs68 cells. Reactive Oxygen Species 26-49 heme oxygenase 1 Homo sapiens 119-135 34940692-7 2021 EEB decreased UVB-induced reactive oxygen species (ROS) generation by upregulating glutathione peroxidase 1 (GPx1) and heme oxygenase-1 (HO-1) expression via nuclear factor erythroid-2-related factor 2 (Nrf2) activation in Hs68 cells. Reactive Oxygen Species 26-49 heme oxygenase 1 Homo sapiens 137-141 34940692-7 2021 EEB decreased UVB-induced reactive oxygen species (ROS) generation by upregulating glutathione peroxidase 1 (GPx1) and heme oxygenase-1 (HO-1) expression via nuclear factor erythroid-2-related factor 2 (Nrf2) activation in Hs68 cells. Reactive Oxygen Species 51-54 heme oxygenase 1 Homo sapiens 119-135 34940692-7 2021 EEB decreased UVB-induced reactive oxygen species (ROS) generation by upregulating glutathione peroxidase 1 (GPx1) and heme oxygenase-1 (HO-1) expression via nuclear factor erythroid-2-related factor 2 (Nrf2) activation in Hs68 cells. Reactive Oxygen Species 51-54 heme oxygenase 1 Homo sapiens 137-141 34634993-1 2021 BACKGROUND: Heme oxygenase (HO)-1 is a rate-limiting enzyme for degrading heme into carbon monoxide. Carbon Monoxide 84-99 heme oxygenase 1 Homo sapiens 12-33 34520103-0 2021 Fenofibrate inhibits hypoxia-inducible factor-1 alpha and carbonic anhydrase expression through activation of AMP-activated protein kinase/HO-1/Sirt1 pathway in glioblastoma cells. Fenofibrate 0-11 heme oxygenase 1 Homo sapiens 139-143 34710668-0 2021 Growing the molecular architecture of imidazole-like ligands in HO-1 complexes. imidazole 38-47 heme oxygenase 1 Homo sapiens 64-68 34520103-7 2021 Moreover, fenofibrate-inhibited HIF-1alpha expression is mediated by HO-1 activation in GBM cells through the AMP-activated protein kinase (AMPK) pathway. Fenofibrate 10-21 heme oxygenase 1 Homo sapiens 69-73 34520103-8 2021 In addition, fenofibrate-enhanced HO-1 upregulation activates SIRT1 and leads to subsequent accumulation of SIRT1 in the nucleus, which further promotes HIF-1alpha deacetylation and inhibits CA9 expression. Fenofibrate 13-24 heme oxygenase 1 Homo sapiens 34-38 34342160-0 2021 Pterostilbene suppresses oxidative stress and allergic airway inflammation through AMPK/Sirt1 and Nrf2/HO-1 pathways. pterostilbene 0-13 heme oxygenase 1 Homo sapiens 103-107 34342160-4 2021 Herein, we aim to explore the pharmacological effects of Pts on oxidative stress and allergic inflammatory response as well as the mechanism involving AMPK/Sirt1 and Nrf2/HO-1 pathways. pterostilbene 57-60 heme oxygenase 1 Homo sapiens 171-175 34633051-0 2021 (Corrigendum) Synergistic cardioprotective effects of Danshensu and hydroxysafflor yellow A against myocardial ischemia-reperfusion injury are mediated through the Akt/Nrf2/HO-1 pathway. danshensu and hydroxysafflor yellow a 54-91 heme oxygenase 1 Homo sapiens 173-177 34167427-0 2021 Novel mutual prodrug of 5-fluorouracil and heme oxygenase-1 inhibitor (5-FU/HO-1 hybrid): design and preliminary in vitro evaluation. Fluorouracil 24-38 heme oxygenase 1 Homo sapiens 43-59 34943103-2 2021 The aim of our study was to assess the role of bilirubin, and the heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variants, which are involved in bilirubin homeostasis, in the NAFLD development in adult patients. Bilirubin 189-198 heme oxygenase 1 Homo sapiens 66-82 34943103-2 2021 The aim of our study was to assess the role of bilirubin, and the heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) promoter gene variants, which are involved in bilirubin homeostasis, in the NAFLD development in adult patients. Bilirubin 189-198 heme oxygenase 1 Homo sapiens 84-89 34943103-5 2021 The (GT)n/(TA)n dinucleotide variations in HMOX1/UGT1A1 gene promoters, respectively, were analyzed by fragment analysis. Nitrogen 8-9 heme oxygenase 1 Homo sapiens 43-48 34943103-5 2021 The (GT)n/(TA)n dinucleotide variations in HMOX1/UGT1A1 gene promoters, respectively, were analyzed by fragment analysis. (ta)n dinucleotide 10-28 heme oxygenase 1 Homo sapiens 43-48 34943105-10 2021 CONCLUSION: In patients with CCS, higher plasma HO-1 levels are associated with lower cholesterol and a more diffuse but mainly non-obstructive coronary atherosclerosis, confirming a potential role for the Nrf2/HO-1 pathway as a protective feedback. Cholesterol 86-97 heme oxygenase 1 Homo sapiens 48-52 34167427-0 2021 Novel mutual prodrug of 5-fluorouracil and heme oxygenase-1 inhibitor (5-FU/HO-1 hybrid): design and preliminary in vitro evaluation. Fluorouracil 24-38 heme oxygenase 1 Homo sapiens 76-80 34167427-0 2021 Novel mutual prodrug of 5-fluorouracil and heme oxygenase-1 inhibitor (5-FU/HO-1 hybrid): design and preliminary in vitro evaluation. Fluorouracil 71-75 heme oxygenase 1 Homo sapiens 43-59 34167427-0 2021 Novel mutual prodrug of 5-fluorouracil and heme oxygenase-1 inhibitor (5-FU/HO-1 hybrid): design and preliminary in vitro evaluation. Fluorouracil 71-75 heme oxygenase 1 Homo sapiens 76-80 34167427-1 2021 In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Fluorouracil 42-56 heme oxygenase 1 Homo sapiens 61-76 34167427-1 2021 In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Fluorouracil 42-56 heme oxygenase 1 Homo sapiens 93-97 34167427-1 2021 In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Fluorouracil 88-92 heme oxygenase 1 Homo sapiens 61-76 34167427-1 2021 In this work, the first mutual prodrug of 5-fluorouracil and heme oxygenase1 inhibitor (5-FU/HO-1 hybrid) has been designed, synthesised, and evaluated for its in vitro chemical and enzymatic hydrolysis stability. Fluorouracil 88-92 heme oxygenase 1 Homo sapiens 93-97 34167427-6 2021 Altogether our results served as an initial proof-of-concept to develop 5-FU/HO-1 mutual prodrugs as potential novel anticancer agents. Fluorouracil 72-76 heme oxygenase 1 Homo sapiens 77-81 34338973-0 2021 Pinocembrin pretreatment counteracts the chlorpyrifos-induced HO-1 downregulation, mitochondrial dysfunction, and inflammation in the SH-SY5Y cells. pinocembrin 0-11 heme oxygenase 1 Homo sapiens 62-66 34663512-3 2021 In this study, the effects of PGG on Nrf2/HO-1 and JAK2/STAT3 signaling were explored in AGE-stimulated mesangial cells. pentagalloylglucose 30-33 heme oxygenase 1 Homo sapiens 42-46 34663512-5 2021 Our results showed that PGG significantly inhibited AGE-induced ROS generation and activated AGE-inhibited Nrf2/HO-1 signaling. pentagalloylglucose 24-27 heme oxygenase 1 Homo sapiens 112-116 34663512-7 2021 Furthermore, ML385 suppressed Nrf2/HO-1 signaling, elevated ROS and cytokine production, and activated JAK2/STAT3 cascade were reversed by PGG. ML385 13-18 heme oxygenase 1 Homo sapiens 35-39 34663512-7 2021 Furthermore, ML385 suppressed Nrf2/HO-1 signaling, elevated ROS and cytokine production, and activated JAK2/STAT3 cascade were reversed by PGG. pentagalloylglucose 139-142 heme oxygenase 1 Homo sapiens 35-39 34663512-8 2021 These findings indicate that PGG inhibits the JAK2/STAT3 cascade by activating Nrf2/HO-1 signaling. pentagalloylglucose 29-32 heme oxygenase 1 Homo sapiens 84-88 34338973-0 2021 Pinocembrin pretreatment counteracts the chlorpyrifos-induced HO-1 downregulation, mitochondrial dysfunction, and inflammation in the SH-SY5Y cells. Chlorpyrifos 41-53 heme oxygenase 1 Homo sapiens 62-66 34338973-8 2021 Silencing of Nrf2 or inhibition of HO-1 suppressed the PB-induced effects in the CPF-challenged cells. pinocembrin 55-57 heme oxygenase 1 Homo sapiens 35-39 34338973-9 2021 Thus, PB promoted beneficial effects by a mechanism dependent on the Nrf2/HO-1/CO + BR axis in the CPF-treated cells. pinocembrin 6-8 heme oxygenase 1 Homo sapiens 74-78 34917622-3 2021 Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Heme 87-91 heme oxygenase 1 Homo sapiens 31-47 34547407-0 2021 Heme Oxygenase-1 (HMOX-1) and inhibitor of differentiation proteins (ID1, ID3) are key response mechanisms against iron-overload in pancreatic beta-cells. Iron 115-119 heme oxygenase 1 Homo sapiens 0-16 34547407-0 2021 Heme Oxygenase-1 (HMOX-1) and inhibitor of differentiation proteins (ID1, ID3) are key response mechanisms against iron-overload in pancreatic beta-cells. Iron 115-119 heme oxygenase 1 Homo sapiens 18-24 34547407-12 2021 Our findings suggest that HMOX1, ID1 and ID3 define the response mechanism against iron-overload-induced stress in beta-cells. Iron 83-87 heme oxygenase 1 Homo sapiens 26-31 34622385-8 2021 The mRNA and protein expression levels of nuclear Nrf2, heme oxygenase 1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1) were significantly increased upon COS treatment. carbonyl sulfide 160-163 heme oxygenase 1 Homo sapiens 56-72 34622385-8 2021 The mRNA and protein expression levels of nuclear Nrf2, heme oxygenase 1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1) were significantly increased upon COS treatment. carbonyl sulfide 160-163 heme oxygenase 1 Homo sapiens 74-78 34622385-9 2021 Moreover, Nrf2-siRNA evidently reversed the promotive effect of COS on expression levels of HO-1 and NQO1, and ARE-driven transcriptional activity as determined by double-luciferase reporter gene assay. carbonyl sulfide 64-67 heme oxygenase 1 Homo sapiens 92-96 34933382-3 2021 HO-1 production can also be stimulated by potassium iodide (KI). Potassium Iodide 42-58 heme oxygenase 1 Homo sapiens 0-4 34933382-3 2021 HO-1 production can also be stimulated by potassium iodide (KI). KS I 60-62 heme oxygenase 1 Homo sapiens 0-4 34933382-8 2021 CONCLUSION: Our findings suggest that KI alleviates oxidative stress leading to decreased HO-1 expression; plasma from preeclamptic women did not induce the enzyme"s expression in HUVECs, and we hypothesize that this is possibly due to inhibitory post-transcriptional mechanisms in response to overexpression of this enzyme during early pregnancy. KS I 39-41 heme oxygenase 1 Homo sapiens 91-95 34600339-12 2021 miR-194 mimic attenuated the effect of OGD/R on cell viability and protein levels of Nrf2, HO-1 and Bach1, whereas Bach1 overexpression reversed the effect of miR-194 mimics. mir-194 0-7 heme oxygenase 1 Homo sapiens 91-95 34876806-0 2021 Methylcobalamin Protects Melanocytes from H2O2-Induced Oxidative Stress by Activating the Nrf2/HO-1 Pathway. mecobalamin 0-15 heme oxygenase 1 Homo sapiens 95-99 34876806-11 2021 Results: We found that MeCbl treatment enhanced cell viability, increased melanin content, reduced intracellular reactive oxygen species (ROS) accumulation, increased the activities of antioxidant enzyme superoxide dismutase (SOD) and catalase (CAT), reduced melanocyte apoptosis, and up-regulated the expression of the Nrf2/HO-1 pathway. mecobalamin 23-28 heme oxygenase 1 Homo sapiens 325-329 34876806-12 2021 Moreover, the protective effects of MeCbl were significantly weakened after inhibiting the Nrf2/HO-1 pathway. mecobalamin 36-41 heme oxygenase 1 Homo sapiens 96-100 34876806-13 2021 Conclusion: Our results indicate that MeCbl attenuated the H2O2-induced oxidative stress in melanocytes by activating the Nrf2/HO-1 pathway, this suggests that MeCbl may be an effective treatment against vitiligo. mecobalamin 38-43 heme oxygenase 1 Homo sapiens 127-131 34876806-13 2021 Conclusion: Our results indicate that MeCbl attenuated the H2O2-induced oxidative stress in melanocytes by activating the Nrf2/HO-1 pathway, this suggests that MeCbl may be an effective treatment against vitiligo. Hydrogen Peroxide 59-63 heme oxygenase 1 Homo sapiens 127-131 34917622-3 2021 Studies have demonstrated that heme oxygenase 1 (HO-1), an inducible enzyme catalyzing heme degradation, exhibits anti-inflammatory, anti-oxidative stress and anti-apoptosis properties. Heme 87-91 heme oxygenase 1 Homo sapiens 49-53 34836523-0 2021 Arctiin suppresses H9N2 avian influenza virus-mediated inflammation via activation of Nrf2/HO-1 signaling. arctiin 0-7 heme oxygenase 1 Homo sapiens 91-95 34836523-11 2021 Interestingly, arctiin treatment obviously reversed H9N2 virus-induced reduction of Nrf2, increased the nuclear translocation of Nrf2, and upregulated Nrf2 signaling target genes (HO-1 and SOD2). arctiin 15-22 heme oxygenase 1 Homo sapiens 180-184 34836523-12 2021 Zinc protoporphyrin (Znpp)-an HO-1 inhibitor-weakened the inhibitory effect of arctiin on H9N2 virus-induced RIG-I/JNK MAPK and proinflammatory mediators. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 30-34 34836523-12 2021 Zinc protoporphyrin (Znpp)-an HO-1 inhibitor-weakened the inhibitory effect of arctiin on H9N2 virus-induced RIG-I/JNK MAPK and proinflammatory mediators. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 30-34 34836523-12 2021 Zinc protoporphyrin (Znpp)-an HO-1 inhibitor-weakened the inhibitory effect of arctiin on H9N2 virus-induced RIG-I/JNK MAPK and proinflammatory mediators. arctiin 79-86 heme oxygenase 1 Homo sapiens 30-34 34836523-13 2021 CONCLUSION: Taken together, these results suggested that the anti-inflammatory effects of arctiin on H9N2 virus infection may be due to the activation of Nrf2/HO-1 and blocked RIG-I/JNK MAPK signaling; thus, arctiin may be a promising agent for prevention and treatment of H9N2 virus infections. arctiin 90-97 heme oxygenase 1 Homo sapiens 159-163 34836523-13 2021 CONCLUSION: Taken together, these results suggested that the anti-inflammatory effects of arctiin on H9N2 virus infection may be due to the activation of Nrf2/HO-1 and blocked RIG-I/JNK MAPK signaling; thus, arctiin may be a promising agent for prevention and treatment of H9N2 virus infections. arctiin 208-215 heme oxygenase 1 Homo sapiens 159-163 34610361-7 2021 Mechanically, our findings revealed that Cd-induced ferroptosis depended upon the excessive activation of Heme oxygenase 1 (HMOX1) and the release of free iron from heme. Cadmium 41-43 heme oxygenase 1 Homo sapiens 106-122 34901124-6 2021 Furthermore, we demonstrated that CS-EVs can alter the gene expression of several genes involved in inflammation (i.e., ICAM1 and HMOX-1) and tight junctions (i.e., OCLN, CLDN1, and MLCK), contributing to limit inflammatory stimuli and restore a functional barrier by increasing the tight junction OCLN protein. CEV regimen 34-40 heme oxygenase 1 Homo sapiens 130-136 34956624-6 2021 Exposure to HCP and ATCP increased both hmox1 and srxn1 gene expression, and was associated with accumulation of Nrf2 protein in cells. hcp 12-15 heme oxygenase 1 Homo sapiens 40-45 34800278-2 2022 HO-1 catalyzes heme breakdown, reducing the levels of this important oxidant molecule and generating antioxidant, anti-inflammatory, and anti-apoptotic byproducts. Heme 15-19 heme oxygenase 1 Homo sapiens 0-4 34610361-7 2021 Mechanically, our findings revealed that Cd-induced ferroptosis depended upon the excessive activation of Heme oxygenase 1 (HMOX1) and the release of free iron from heme. Cadmium 41-43 heme oxygenase 1 Homo sapiens 124-129 34610361-9 2021 Collectively, Cd induced ferroptosis by iron homeostasis dysregulation, mediated by excessive activation of HMOX-1. Cadmium 14-16 heme oxygenase 1 Homo sapiens 108-114 34562468-0 2021 Astragalus polysaccharide prevents ferroptosis in a murine model of experimental colitis and human Caco-2 cells via inhibiting NRF2/HO-1 pathway. Polysaccharides 11-25 heme oxygenase 1 Homo sapiens 132-136 34840667-10 2021 Moreover, inhibition of HO-1 by ZnPP weakened the cytoprotective effect of NAR. zinc protoporphyrin 32-36 heme oxygenase 1 Homo sapiens 24-28 34840667-10 2021 Moreover, inhibition of HO-1 by ZnPP weakened the cytoprotective effect of NAR. naringenin 75-78 heme oxygenase 1 Homo sapiens 24-28 34867473-11 2021 Moreover, melatonin can repair the endothelial damage from preeclampsia at the placenta level, increasing PIGF, Nrf-2, HO-1 production and reducing critical markers of vascular injury during the pregnancy. Melatonin 10-19 heme oxygenase 1 Homo sapiens 119-123 34787456-0 2022 The Antimalaria Drug Artesunate Inhibits Porcine Reproductive and Respiratory Syndrome Virus Replication via Activating AMPK and Nrf2/HO-1 Signaling Pathways. Artesunate 21-31 heme oxygenase 1 Homo sapiens 134-138 34582846-5 2021 In particular, this biomolecule reduces the generation of reactive oxygen species (ROS) by blocking the activation of inflammatory transcription factors, especially nuclear factor-kappaB (NF-kappaB), and accelerating the synthesis of antioxidant compounds such as heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). Reactive Oxygen Species 83-86 heme oxygenase 1 Homo sapiens 264-280 34582846-5 2021 In particular, this biomolecule reduces the generation of reactive oxygen species (ROS) by blocking the activation of inflammatory transcription factors, especially nuclear factor-kappaB (NF-kappaB), and accelerating the synthesis of antioxidant compounds such as heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). Reactive Oxygen Species 83-86 heme oxygenase 1 Homo sapiens 282-286 34582846-5 2021 In particular, this biomolecule reduces the generation of reactive oxygen species (ROS) by blocking the activation of inflammatory transcription factors, especially nuclear factor-kappaB (NF-kappaB), and accelerating the synthesis of antioxidant compounds such as heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 264-280 34582846-5 2021 In particular, this biomolecule reduces the generation of reactive oxygen species (ROS) by blocking the activation of inflammatory transcription factors, especially nuclear factor-kappaB (NF-kappaB), and accelerating the synthesis of antioxidant compounds such as heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 282-286 34858164-4 2021 It is showed that some natural compounds such as flavonoids, phenols, glycosides and coumarins have a protective role in melanocytes and thereby arrest the depigmentation, and, additionally, Nrf2/HO-1, MAPK, JAK/STAT, cAMP/PKA, and Wnt/beta-catenin signaling pathways were reported to be implicated in these protective effects. Phenols 61-68 heme oxygenase 1 Homo sapiens 196-200 34782602-0 2021 Iron overload in endometriosis peritoneal fluid induces early embryo ferroptosis mediated by HMOX1. Iron 0-4 heme oxygenase 1 Homo sapiens 93-98 34858164-4 2021 It is showed that some natural compounds such as flavonoids, phenols, glycosides and coumarins have a protective role in melanocytes and thereby arrest the depigmentation, and, additionally, Nrf2/HO-1, MAPK, JAK/STAT, cAMP/PKA, and Wnt/beta-catenin signaling pathways were reported to be implicated in these protective effects. Glycosides 70-80 heme oxygenase 1 Homo sapiens 196-200 34858164-4 2021 It is showed that some natural compounds such as flavonoids, phenols, glycosides and coumarins have a protective role in melanocytes and thereby arrest the depigmentation, and, additionally, Nrf2/HO-1, MAPK, JAK/STAT, cAMP/PKA, and Wnt/beta-catenin signaling pathways were reported to be implicated in these protective effects. Flavonoids 49-59 heme oxygenase 1 Homo sapiens 196-200 34858164-4 2021 It is showed that some natural compounds such as flavonoids, phenols, glycosides and coumarins have a protective role in melanocytes and thereby arrest the depigmentation, and, additionally, Nrf2/HO-1, MAPK, JAK/STAT, cAMP/PKA, and Wnt/beta-catenin signaling pathways were reported to be implicated in these protective effects. Coumarins 85-94 heme oxygenase 1 Homo sapiens 196-200 34803685-0 2021 Paeoniflorin Suppresses TBHP-Induced Oxidative Stress and Apoptosis in Human Umbilical Vein Endothelial Cells via the Nrf2/HO-1 Signaling Pathway and Improves Skin Flap Survival. peoniflorin 0-12 heme oxygenase 1 Homo sapiens 123-127 34803685-11 2021 However, the PF-mediated protection was partially lost after co-treatment with ML385, a selective Nrf2 inhibitor, suggesting that PF is a crucial modulator regulating the Nrf2/HO-1 signaling pathway. ML385 79-84 heme oxygenase 1 Homo sapiens 176-180 34478827-7 2021 More so, treatment with ZnPP under hypoxic conditions down-regulated HO-1 expression, stimulated proliferation, and resisted apoptosis in yak PASMCs. zinc protoporphyrin 24-28 heme oxygenase 1 Homo sapiens 69-73 34728736-0 2021 Hemin as a novel candidate for treating COVID-19 via heme oxygenase-1 induction. Hemin 0-5 heme oxygenase 1 Homo sapiens 53-69 34728736-5 2021 To confirm whether HO-1 suppresses SARS-CoV-2 infection, we assessed the antiviral activity of hemin, an effective and safe HO-1 inducer, in SARS-CoV-2 infection. Hemin 95-100 heme oxygenase 1 Homo sapiens 124-128 34728736-8 2021 Free iron and biliverdin, which are metabolic byproducts of heme catalysis by HO-1, also suppressed the viral infection. Iron 5-9 heme oxygenase 1 Homo sapiens 78-82 34728736-8 2021 Free iron and biliverdin, which are metabolic byproducts of heme catalysis by HO-1, also suppressed the viral infection. Biliverdine 14-24 heme oxygenase 1 Homo sapiens 78-82 34728736-8 2021 Free iron and biliverdin, which are metabolic byproducts of heme catalysis by HO-1, also suppressed the viral infection. Heme 60-64 heme oxygenase 1 Homo sapiens 78-82 34728736-10 2021 Overall, the findings suggested that HO-1, induced by hemin, effectively suppressed SARS-CoV-2 in vitro. Hemin 54-59 heme oxygenase 1 Homo sapiens 37-41 34796171-0 2021 Butyric Acid Protects Against Renal Ischemia-Reperfusion Injury by Adjusting the Treg/Th17 Balance via HO-1/p-STAT3 Signaling. Butyric Acid 0-12 heme oxygenase 1 Homo sapiens 103-107 34796171-0 2021 Butyric Acid Protects Against Renal Ischemia-Reperfusion Injury by Adjusting the Treg/Th17 Balance via HO-1/p-STAT3 Signaling. treg 81-85 heme oxygenase 1 Homo sapiens 103-107 34796171-5 2021 Mechanism studies have shown that heme oxygenase 1 (HO-1)/STAT3 signaling pathway was involved in the inhibitory effect of BA on Th17 cell differentiation. Butyric Acid 123-125 heme oxygenase 1 Homo sapiens 34-50 34796171-5 2021 Mechanism studies have shown that heme oxygenase 1 (HO-1)/STAT3 signaling pathway was involved in the inhibitory effect of BA on Th17 cell differentiation. Butyric Acid 123-125 heme oxygenase 1 Homo sapiens 52-56 34796171-6 2021 HO-1 inhibitors can significantly rescue the BA-mediated inhibition of Th17 cell differentiation. Butyric Acid 45-47 heme oxygenase 1 Homo sapiens 0-4 34510844-0 2021 Echinatin mitigates H2O2-induced oxidative damage and apoptosis in lens epithelial cells via the Nrf2/HO-1 pathway. echinatin 0-9 heme oxygenase 1 Homo sapiens 102-106 34510844-10 2021 Moreover, H2O2 significantly reduced Nrf2 nuclear localization, as well as HO-1 and NQO1 expression, which could be reversed by Ech. Hydrogen Peroxide 10-14 heme oxygenase 1 Homo sapiens 75-79 34510844-12 2021 CONCLUSIONS: Echinatin mitigates H2O2-induced oxidative damage and apoptosis in HLECs via the Nrf2/HO-1 pathway, suggesting that Ech may be a potential drug for the treatment of cataracts. echinatin 13-22 heme oxygenase 1 Homo sapiens 99-103 34298225-0 2021 Paeoniflorin ameliorates oxidase stress in Glutamate-stimulated SY5Y and prenatally stressed female offspring through Nrf2/HO-1 signaling pathway. peoniflorin 0-12 heme oxygenase 1 Homo sapiens 123-127 34146326-10 2021 Interestingly, melatonin not only markedly decreased the protein levels of TRPV1 and CGRP, but also enhanced the expression of Nrf2, quinone oxidoreductase-1 (NQO-1), and heme oxygenase-1 (HO-1). Melatonin 15-24 heme oxygenase 1 Homo sapiens 171-187 34146326-10 2021 Interestingly, melatonin not only markedly decreased the protein levels of TRPV1 and CGRP, but also enhanced the expression of Nrf2, quinone oxidoreductase-1 (NQO-1), and heme oxygenase-1 (HO-1). Melatonin 15-24 heme oxygenase 1 Homo sapiens 189-193 34582963-3 2021 Western blot analysis indicated that C3G and PCA minimized PhIP-induced cell damage by reversing the abnormal expression of Bax/Bcl-2, Cytochrome c, cleaved Caspase-3, XIAP, Nrf2, HO-1, LC3 and p62 involved in intrinsic apoptotic and Nrf2/p62 pathways. 2-amino-1-methyl-6-phenylimidazo(4,5-b)pyridine 59-63 heme oxygenase 1 Homo sapiens 180-184 34769290-9 2021 Data from the Human Apoptosis Array revealed that FLLL32 increases the expression of cleaved caspase-3 and heme oxygenase-1 (HO-1). FLLL 32 50-56 heme oxygenase 1 Homo sapiens 107-123 34769290-9 2021 Data from the Human Apoptosis Array revealed that FLLL32 increases the expression of cleaved caspase-3 and heme oxygenase-1 (HO-1). FLLL 32 50-56 heme oxygenase 1 Homo sapiens 125-129 34769290-12 2021 In conclusion, our findings suggest that FLLL32 is a potential therapeutic agent for oral cancer by inducing caspase-dependent apoptosis and HO-1 activation through the p38 pathway. FLLL 32 41-47 heme oxygenase 1 Homo sapiens 141-145 34822480-5 2021 The cytoprotective mechanism of these peptides involves an improvement in the cellular antioxidant defense system, as indicated by the suppression of the intracellular ROS generation through upregulation of the cytoprotective enzyme heme oxygenase-1. ros 168-171 heme oxygenase 1 Homo sapiens 233-249 34769290-13 2021 We believe that the activation of HO-1 and the p38 pathway by FLLL32 represent potential targets for further research in oral cancer. FLLL 32 62-68 heme oxygenase 1 Homo sapiens 34-38 34508760-9 2021 More importantly, HMOX1 knockdown attenuated Fe2+ overload, reduced iron content and ROS, and alleviated lipid peroxidation, which led to a reduction in ferroptosis in diabetic human endothelial cells. ammonium ferrous sulfate 45-49 heme oxygenase 1 Homo sapiens 18-23 34508760-9 2021 More importantly, HMOX1 knockdown attenuated Fe2+ overload, reduced iron content and ROS, and alleviated lipid peroxidation, which led to a reduction in ferroptosis in diabetic human endothelial cells. Iron 68-72 heme oxygenase 1 Homo sapiens 18-23 34508760-9 2021 More importantly, HMOX1 knockdown attenuated Fe2+ overload, reduced iron content and ROS, and alleviated lipid peroxidation, which led to a reduction in ferroptosis in diabetic human endothelial cells. ros 85-88 heme oxygenase 1 Homo sapiens 18-23 34558653-0 2021 Ginsenoside Rk1 protects human melanocytes from H2O2-induced oxidative injury via regulation of the PI3K/AKT/Nrf2/HO-1 pathway. ginsenoside Rk1 0-15 heme oxygenase 1 Homo sapiens 114-118 34558653-11 2021 In addition, RK1 treatment notably induced Nrf2 nuclear translocation, increased the protein expression levels of Nrf2 and HO-1, and the ratio of phosphorylated-AKT to AKT in the PIG1 cells exposed to H2O2. Hydrogen Peroxide 201-205 heme oxygenase 1 Homo sapiens 123-127 34754326-11 2021 Conclusion: Our data suggested that treatment with DMF mitigated LPS-induced oxidative stress, inflammation, and ER stress-associated apoptosis in NPCs via the Nrf2/HO-1 signaling pathway, thus reliving LPS-induced dysfunction of NPCs, which offered a novel potential pharmacological treatment strategy for IVDD. Dimethyl Fumarate 51-54 heme oxygenase 1 Homo sapiens 165-169 34737578-6 2021 Hydrogen sulfide (H2S) likewise opposes mast cell activation; H2S can boost AMPK activity, up-regulate cGMP production, and trigger Nrf2-mediated induction of Phase 2 enzymes - including heme oxygenase-1, whose generation of bilirubin suppresses NADPH oxidase activity. Deuterium 18-21 heme oxygenase 1 Homo sapiens 187-203 34737578-6 2021 Hydrogen sulfide (H2S) likewise opposes mast cell activation; H2S can boost AMPK activity, up-regulate cGMP production, and trigger Nrf2-mediated induction of Phase 2 enzymes - including heme oxygenase-1, whose generation of bilirubin suppresses NADPH oxidase activity. Deuterium 62-65 heme oxygenase 1 Homo sapiens 187-203 34737578-6 2021 Hydrogen sulfide (H2S) likewise opposes mast cell activation; H2S can boost AMPK activity, up-regulate cGMP production, and trigger Nrf2-mediated induction of Phase 2 enzymes - including heme oxygenase-1, whose generation of bilirubin suppresses NADPH oxidase activity. Bilirubin 225-234 heme oxygenase 1 Homo sapiens 187-203 34509368-6 2021 In summary, WEKPPVSH might protect against oxidative stress and inflammation in LPS-stimulated BV-2 microglia by enhancing the Nrf2/HO-1 signaling pathway and blocking the nuclear factor-kappaB/p38 mitogen - activated protein kinase (NF-kappaB/p38 MAPK) signaling pathway. wekppvsh 12-20 heme oxygenase 1 Homo sapiens 132-136 34609010-7 2021 Our results showed that berberine-LCNs significantly reduced the expression of CCl-20, CXCL-8 and HO-1 at dose of 5microM. Berberine 24-33 heme oxygenase 1 Homo sapiens 98-102 34609010-7 2021 Our results showed that berberine-LCNs significantly reduced the expression of CCl-20, CXCL-8 and HO-1 at dose of 5microM. lcns 34-38 heme oxygenase 1 Homo sapiens 98-102 34609010-8 2021 Collectively, our findings suggest that berberine-LCNs have inhibitory effect on inflammation/oxidative stress related cytokines i.e. CCL20, CXCL-8, and HO-1 which could be a novel therapeutic target for the management of lung cancer. Berberine 40-49 heme oxygenase 1 Homo sapiens 153-157 34609010-11 2021 Our study showed that Berberine LCNs significantly downregulate the expression of CXCL-8, CCL-20 and HO-1 which suggests that Berberine loaded nanoparticles could be a promising therapeutic alternative for the management of lung cancer. berberine lcns 22-36 heme oxygenase 1 Homo sapiens 101-105 34609010-11 2021 Our study showed that Berberine LCNs significantly downregulate the expression of CXCL-8, CCL-20 and HO-1 which suggests that Berberine loaded nanoparticles could be a promising therapeutic alternative for the management of lung cancer. Berberine 126-135 heme oxygenase 1 Homo sapiens 101-105 34453944-1 2021 AIM: Biliverdin reductase-A (BVR-A) other than its canonical role in the degradation pathway of heme as partner of heme oxygenase-1 (HO1), has recently drawn attention as a protein with pleiotropic functions involved in insulin-glucose homeostasis. Heme 96-100 heme oxygenase 1 Homo sapiens 115-131 34453944-1 2021 AIM: Biliverdin reductase-A (BVR-A) other than its canonical role in the degradation pathway of heme as partner of heme oxygenase-1 (HO1), has recently drawn attention as a protein with pleiotropic functions involved in insulin-glucose homeostasis. Heme 96-100 heme oxygenase 1 Homo sapiens 133-136 34476501-4 2021 The expression levels of apoptosis-related proteins, activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway induced by H2O2 and the effect of treatment with ANP on vertebral EPCs were detected by western blotting. Hydrogen Peroxide 175-179 heme oxygenase 1 Homo sapiens 122-138 34476501-4 2021 The expression levels of apoptosis-related proteins, activation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway induced by H2O2 and the effect of treatment with ANP on vertebral EPCs were detected by western blotting. Hydrogen Peroxide 175-179 heme oxygenase 1 Homo sapiens 140-144 34476501-8 2021 Finally, ANP blocked H2O2-induced oxidative stress through activating the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 21-25 heme oxygenase 1 Homo sapiens 79-83 34476501-9 2021 These findings suggested that ANP may effectively protect EPCs through inhibition of H2O2-induced oxidant injury and cell death by activating the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 85-89 heme oxygenase 1 Homo sapiens 151-155 34688156-2 2021 With the continuous deepening of research, in addition to directly regulating redox by catalyzing the degradation of heme, HO-1 protein also participates in the gene expression level in a great diversity of methods, thereby initiating cell defense. Heme 117-121 heme oxygenase 1 Homo sapiens 123-127 34688156-5 2021 Especially for malignant diseases, there may be new advances in the treatment of HO-1 by regulating abnormal ROS and metabolic signaling. ros 109-112 heme oxygenase 1 Homo sapiens 81-85 34754326-0 2021 Dimethyl Fumarate Ameliorates Nucleus Pulposus Cell Dysfunction through Activating the Nrf2/HO-1 Pathway in Intervertebral Disc Degeneration. Dimethyl Fumarate 0-17 heme oxygenase 1 Homo sapiens 92-96 34737578-6 2021 Hydrogen sulfide (H2S) likewise opposes mast cell activation; H2S can boost AMPK activity, up-regulate cGMP production, and trigger Nrf2-mediated induction of Phase 2 enzymes - including heme oxygenase-1, whose generation of bilirubin suppresses NADPH oxidase activity. Hydrogen Sulfide 0-16 heme oxygenase 1 Homo sapiens 187-203 34269613-5 2021 Internalization of ferrylHb is accompanied by up-regulation of heme oxygenase-1 and H-ferritin and accumulation of iron within lysosomes as a result of heme/iron uptake. Heme 152-156 heme oxygenase 1 Homo sapiens 63-79 34828827-0 2021 Selenium-Enriched Soy Protein Has Antioxidant Potential via Modulation of the NRF2-HO1 Signaling Pathway. Selenium 0-8 heme oxygenase 1 Homo sapiens 83-86 34768836-0 2021 5-Aminolevulinic Acid Attenuates Glucose-Regulated Protein 78 Expression and Hepatocyte Lipoapoptosis via Heme Oxygenase-1 Induction. 5-amino levulinic acid 0-21 heme oxygenase 1 Homo sapiens 106-122 34768836-12 2021 5-ALA significantly induced HO-1 and decreased GRP78 expression. 5-amino levulinic acid 0-5 heme oxygenase 1 Homo sapiens 28-32 34768836-16 2021 5-ALA has a therapeutic potential on hepatic steatosis by suppressing ER stress and lipoapoptosis by attenuating GRP78 via HO-1 induction. 5-amino levulinic acid 0-5 heme oxygenase 1 Homo sapiens 123-127 34269613-5 2021 Internalization of ferrylHb is accompanied by up-regulation of heme oxygenase-1 and H-ferritin and accumulation of iron within lysosomes as a result of heme/iron uptake. Iron 157-161 heme oxygenase 1 Homo sapiens 63-79 34622673-10 2021 In eAAA, HO-1 expression was negatively correlated with aortic collagen content and oxidative stress parameters H2O2 release, oxidized proteins, and thiobarbituric acid reactive substances. Hydrogen Peroxide 112-116 heme oxygenase 1 Homo sapiens 9-13 34622673-10 2021 In eAAA, HO-1 expression was negatively correlated with aortic collagen content and oxidative stress parameters H2O2 release, oxidized proteins, and thiobarbituric acid reactive substances. thiobarbituric acid 149-168 heme oxygenase 1 Homo sapiens 9-13 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. Bexarotene 111-121 heme oxygenase 1 Homo sapiens 169-174 34679025-0 2021 The Protective Effect of Heme Oxygenase-1 on Liver Injury Caused by DON-Induced Oxidative Stress and Cytotoxicity. deoxynivalenol 68-71 heme oxygenase 1 Homo sapiens 25-41 34679025-10 2021 Significantly, AAV-mediated liver-specific overexpression of HO-1 reduced DON-induced liver damage and indirectly protected the abilities of antioxidant enzyme/DNA damage repair system, while AAV-mediated silence of HO-1 produced the opposite effect. deoxynivalenol 74-77 heme oxygenase 1 Homo sapiens 61-65 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. Bexarotene 111-121 heme oxygenase 1 Homo sapiens 314-319 34679025-11 2021 In addition, we found that overexpression of HO-1 could enhance autophagy and combined it with an antioxidant enzyme/DNA damage repair system to inhibit DON-induced hepatocyte damage. deoxynivalenol 153-156 heme oxygenase 1 Homo sapiens 45-49 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. desloratadine 127-140 heme oxygenase 1 Homo sapiens 169-174 34679025-12 2021 Altogether, these data suggest that HO-1 reduces the oxidative stress and DNA damage caused by DON sub-chronic exposure through maintaining DNA repair, antioxidant activity, as well as autophagy. deoxynivalenol 95-98 heme oxygenase 1 Homo sapiens 36-40 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. desloratadine 127-140 heme oxygenase 1 Homo sapiens 314-319 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. conivaptan 99-109 heme oxygenase 1 Homo sapiens 169-174 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. 1-(adamantan-1-yl)-2-(1h-imidazol-1-yl)ethenone 226-273 heme oxygenase 1 Homo sapiens 169-174 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. conivaptan 99-109 heme oxygenase 1 Homo sapiens 314-319 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. 1-(adamantan-1-yl)-2-(1h-imidazol-1-yl)ethenone 226-273 heme oxygenase 1 Homo sapiens 314-319 34681275-6 2021 Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. Ellagic Acid 278-290 heme oxygenase 1 Homo sapiens 314-319 34681275-7 2021 Conivaptan treatment increased doxorubicin sensitivity of both HMOX1- and PRKCA-transfected cell lines. conivaptan 0-10 heme oxygenase 1 Homo sapiens 63-68 34119834-7 2021 In an LPS-induced peritoneal macrophage cell model, this compound could cause a significant increase in the nuclear Nrf2 protein, decrease in the cytosolic Nrf2 protein, and further elevate the downstream protective enzymes HO-1 and NQO-1, which were better than the lead compound NXPZ-2 and sulforaphane. nxpz-2 281-287 heme oxygenase 1 Homo sapiens 224-228 34681275-8 2021 Bexarotene treatment had a comparable doxorubicin-sensitizing effect in HMOX1-transfected cells and desloratadine in PRKCA-transfected cells. Bexarotene 0-10 heme oxygenase 1 Homo sapiens 72-77 34119834-7 2021 In an LPS-induced peritoneal macrophage cell model, this compound could cause a significant increase in the nuclear Nrf2 protein, decrease in the cytosolic Nrf2 protein, and further elevate the downstream protective enzymes HO-1 and NQO-1, which were better than the lead compound NXPZ-2 and sulforaphane. sulforaphane 292-304 heme oxygenase 1 Homo sapiens 224-228 34190424-3 2021 To enhance the efficacy of PDT for tumor treatment, a versatile nanoparticle (NP)-based drug is developed, which the authors call PZB NP, containing the glutathione inhibitor l-buthionine sulfoximine (BSO) and the heme oxygenase 1 (HO-1) inhibitor protoporphyrin zinc(II) (ZnPP) to suppress the innate antioxidant defense system of cancer cells in a two-pronged manner. protoporphyrin zinc(ii) 248-271 heme oxygenase 1 Homo sapiens 214-230 34681608-0 2021 Bisphenol a Induces Autophagy Defects and AIF-Dependent Apoptosis via HO-1 and AMPK to Degenerate N2a Neurons. bis(4-hydroxyphenyl)sulfone 0-9 heme oxygenase 1 Homo sapiens 70-74 34680118-0 2021 Protocatechuic Acid, a Simple Plant Secondary Metabolite, Induced Apoptosis by Promoting Oxidative Stress through HO-1 Downregulation and p21 Upregulation in Colon Cancer Cells. protocatechuic acid 0-19 heme oxygenase 1 Homo sapiens 114-118 34265290-8 2021 Inhibition of the enzyme heme oxygenase-1 (HO-1) or silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) suppressed the AST-promoted cellular and mitochondrial protection. astaxanthine 156-159 heme oxygenase 1 Homo sapiens 25-41 34265290-8 2021 Inhibition of the enzyme heme oxygenase-1 (HO-1) or silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) suppressed the AST-promoted cellular and mitochondrial protection. astaxanthine 156-159 heme oxygenase 1 Homo sapiens 43-47 34681608-8 2021 Both heme oxygenase (HO)-1 and AMP-activated protein kinase (AMPK) up-regulated/activated by BPA mediated the molecular signalings involved in apoptosis and autophagy. bisphenol A 93-96 heme oxygenase 1 Homo sapiens 5-26 34681608-9 2021 HO-1 inhibition or AIF silencing effectively reduced BPA-induced neuronal death. bisphenol A 53-56 heme oxygenase 1 Homo sapiens 0-4 34681608-11 2021 These results suggest that BPA induces N2a neurotoxicity characterized by AIF-dependent apoptosis and p62-related autophagy defects via HO-1 up-regulation and AMPK activation, thereby resulting in neuronal degeneration. bisphenol A 27-30 heme oxygenase 1 Homo sapiens 136-140 34680118-7 2021 The phenolic acid induced modifications in levels of HO-1, non-proteic thiol groups, gamma-glutamylcysteine synthetase, reactive oxygen species, and p21. phenolic acid 4-17 heme oxygenase 1 Homo sapiens 53-57 34265290-10 2021 Overall, our data demonstrate that AST prevented mitochondrial dysfunction by a mechanism related to the Nrf2/HO-1 axis in GLU-challenged cells. Glutamic Acid 123-126 heme oxygenase 1 Homo sapiens 110-114 34190424-3 2021 To enhance the efficacy of PDT for tumor treatment, a versatile nanoparticle (NP)-based drug is developed, which the authors call PZB NP, containing the glutathione inhibitor l-buthionine sulfoximine (BSO) and the heme oxygenase 1 (HO-1) inhibitor protoporphyrin zinc(II) (ZnPP) to suppress the innate antioxidant defense system of cancer cells in a two-pronged manner. protoporphyrin zinc(ii) 248-271 heme oxygenase 1 Homo sapiens 232-236 34190424-3 2021 To enhance the efficacy of PDT for tumor treatment, a versatile nanoparticle (NP)-based drug is developed, which the authors call PZB NP, containing the glutathione inhibitor l-buthionine sulfoximine (BSO) and the heme oxygenase 1 (HO-1) inhibitor protoporphyrin zinc(II) (ZnPP) to suppress the innate antioxidant defense system of cancer cells in a two-pronged manner. zinc protoporphyrin 273-277 heme oxygenase 1 Homo sapiens 214-230 34190424-3 2021 To enhance the efficacy of PDT for tumor treatment, a versatile nanoparticle (NP)-based drug is developed, which the authors call PZB NP, containing the glutathione inhibitor l-buthionine sulfoximine (BSO) and the heme oxygenase 1 (HO-1) inhibitor protoporphyrin zinc(II) (ZnPP) to suppress the innate antioxidant defense system of cancer cells in a two-pronged manner. zinc protoporphyrin 273-277 heme oxygenase 1 Homo sapiens 232-236 34190424-4 2021 BSO reduces intracellular glutathione levels to minimize ROS elimination and protein protection during PDT, and ZnPP inhibits the ROS-stimulated upregulation of the antioxidant HO-1, thus preventing ROS removal by cells after PDT. zinc protoporphyrin 112-116 heme oxygenase 1 Homo sapiens 177-181 34190424-4 2021 BSO reduces intracellular glutathione levels to minimize ROS elimination and protein protection during PDT, and ZnPP inhibits the ROS-stimulated upregulation of the antioxidant HO-1, thus preventing ROS removal by cells after PDT. ros 130-133 heme oxygenase 1 Homo sapiens 177-181 34190424-4 2021 BSO reduces intracellular glutathione levels to minimize ROS elimination and protein protection during PDT, and ZnPP inhibits the ROS-stimulated upregulation of the antioxidant HO-1, thus preventing ROS removal by cells after PDT. ros 199-202 heme oxygenase 1 Homo sapiens 177-181 34480604-5 2021 Bardoxolone-methyl (CDDO-Me) induced oxidative stress caused similarly increased expression of both the wild-type and eGFP-tagged HMOX1 at the mRNA and protein level. bardoxolone methyl 0-18 heme oxygenase 1 Homo sapiens 130-135 34293961-10 2021 Meanwhile, administration of antioxidants or heme oxygenase-1 induction to elevate the hepatocellular levels of the endogenous scavenger bilirubin are promising alternatives that need to be re-evaluated and implemented. Bilirubin 137-146 heme oxygenase 1 Homo sapiens 45-61 34480604-5 2021 Bardoxolone-methyl (CDDO-Me) induced oxidative stress caused similarly increased expression of both the wild-type and eGFP-tagged HMOX1 at the mRNA and protein level. bardoxolone methyl 20-27 heme oxygenase 1 Homo sapiens 130-135 34189835-5 2021 Pb nanoparticles also induced the gene expression of heme oxygenase-1 in dTHP-1 cells but not in A549 cells. Lead 0-2 heme oxygenase 1 Homo sapiens 53-69 34346143-0 2021 Arbutin attenuates ethanol-induced acute hepatic injury by the modulation of oxidative stress and Nrf-2/HO-1 signaling pathway. Ethanol 19-26 heme oxygenase 1 Homo sapiens 104-108 34380011-5 2021 The QUE was also found to counteract STE-induced ROS generation and decrease STE-induced up-regulation of the expression of the stress-inducible enzymes HO-1 and NOS-2. Sterigmatocystin 77-80 heme oxygenase 1 Homo sapiens 153-157 34346143-12 2021 Arbutin triggered Nrf-2/HO-1 signaling cascade liver tissues of ethanol-provoked animals. Arbutin 0-7 heme oxygenase 1 Homo sapiens 24-28 34346143-0 2021 Arbutin attenuates ethanol-induced acute hepatic injury by the modulation of oxidative stress and Nrf-2/HO-1 signaling pathway. Arbutin 0-7 heme oxygenase 1 Homo sapiens 104-108 34346143-12 2021 Arbutin triggered Nrf-2/HO-1 signaling cascade liver tissues of ethanol-provoked animals. Ethanol 64-71 heme oxygenase 1 Homo sapiens 24-28 34487292-9 2021 The intracellular antioxidant enzymes HO-1 and SOD-2 were upregulated by neoxanthin pretreatment. neoxanthin 73-83 heme oxygenase 1 Homo sapiens 38-42 34241977-0 2021 Computational Designing and Prediction of ADMET Properties of Four Novel Imidazole-Based Drug Candidates Inhibiting Heme Oxygenase-1 Causing Cancers. imidazole 73-82 heme oxygenase 1 Homo sapiens 116-132 34241977-2 2021 The inhibition of HO-1 through imidazole-based drugs, which is non-competitive with heme, is a focus of anticancer drug research. imidazole 31-40 heme oxygenase 1 Homo sapiens 18-22 34241977-2 2021 The inhibition of HO-1 through imidazole-based drugs, which is non-competitive with heme, is a focus of anticancer drug research. Heme 84-88 heme oxygenase 1 Homo sapiens 18-22 34530349-4 2021 In the present study, we found that iron overload promoted ferroptosis in hepatocytes by excessively inducing HO-1 expression, which contributed to the progression of liver injury and fibrosis, accompanied by the upregulation of the FGF21 protein level in vitro and in vivo. Iron 36-40 heme oxygenase 1 Homo sapiens 110-114 34470469-0 2021 The protective benefit of heme oxygenase-1 gene modified human placenta-derived Mesenchymal stem cell in N-nitro-L-arginine methyl ester -induced preeclampsia-like rat model: Possible implications for placental angiogenesis. n-nitro-l-arginine methyl ester 105-136 heme oxygenase 1 Homo sapiens 26-42 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. cu-hemin-peg-la 38-53 heme oxygenase 1 Homo sapiens 210-226 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. cu-hemin-peg-la 38-53 heme oxygenase 1 Homo sapiens 228-233 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. weak acid 81-90 heme oxygenase 1 Homo sapiens 210-226 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. weak acid 81-90 heme oxygenase 1 Homo sapiens 228-233 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. cu(ii) 103-109 heme oxygenase 1 Homo sapiens 210-226 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. cu(ii) 103-109 heme oxygenase 1 Homo sapiens 228-233 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. Hemin 114-119 heme oxygenase 1 Homo sapiens 210-226 34448349-4 2021 After highly ingested by HepG2 cells, Cu-Hemin-PEG-LA nanosheets are degraded by weak acid and release Cu(II) and hemin, which consuming intracellular glutathione (GSH) content and increasing the expression of heme oxygenase 1 (HMOX1) protein, respectively. Hemin 114-119 heme oxygenase 1 Homo sapiens 228-233 34448349-6 2021 The intracellular Fe2+ content is overloaded by the significant up-regulation of HMOX1 expression, which is denoted as nonclassical mode. ammonium ferrous sulfate 18-22 heme oxygenase 1 Homo sapiens 81-86 34583348-3 2021 Erythrocyte destruction results in increased release of cytotoxic free heme that is scavenged by haptoglobin (Hp), hemopexin (Hx) and heme oxygenase-1 (HO-1). Heme 71-75 heme oxygenase 1 Homo sapiens 134-150 34089821-1 2021 The intracellular enzyme heme oxygenase-1 (HO-1) is responsible for the degradation of cell-free (cf) heme. Heme 102-106 heme oxygenase 1 Homo sapiens 25-41 34089821-1 2021 The intracellular enzyme heme oxygenase-1 (HO-1) is responsible for the degradation of cell-free (cf) heme. Heme 102-106 heme oxygenase 1 Homo sapiens 43-47 34583348-3 2021 Erythrocyte destruction results in increased release of cytotoxic free heme that is scavenged by haptoglobin (Hp), hemopexin (Hx) and heme oxygenase-1 (HO-1). Heme 71-75 heme oxygenase 1 Homo sapiens 152-156 34583348-7 2021 A cross-sectional study was conducted to determine the association between plasma levels of free heme, HO-1, Hp, Hx, and malaria status in pregnant women who received routine iron supplementation and their birth outcomes. Iron 175-179 heme oxygenase 1 Homo sapiens 103-107 34472337-0 2021 Discovery of Novel Acetamide-Based Heme Oxygenase-1 Inhibitors with Potent In Vitro Antiproliferative Activity. acetamide 19-28 heme oxygenase 1 Homo sapiens 35-51 34472337-1 2021 Heme oxygenase-1 (HO-1) promotes heme catabolism exercising cytoprotective roles in normal and cancer cells. Heme 33-37 heme oxygenase 1 Homo sapiens 0-16 34472337-1 2021 Heme oxygenase-1 (HO-1) promotes heme catabolism exercising cytoprotective roles in normal and cancer cells. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 34638586-13 2021 The upregulation of NEDD4-1 attenuated PTEN expression and promoted the AKT/NRF2/HO-1 oxidative stress signaling axis, which in turn conferred amplified defense against reactive oxygen species (ROS) and eventually higher resistance against TMZ treatment. Reactive Oxygen Species 169-192 heme oxygenase 1 Homo sapiens 81-85 34630071-7 2021 Mechanistically, the expression of NADPH oxidase 1 (Nox1) was found to be suppressed and antioxidant enzymes including catalase, superoxide dismutase 2 (SOD-2), and heme oxygenase-1(HO-1) were enhanced following RH treatment, suggesting RH exhibited antioxidant activity by reducing the generation of reactive oxygen species (ROS) as well as enhancing the depletion of ROS. rhapontin 212-214 heme oxygenase 1 Homo sapiens 165-181 34630071-7 2021 Mechanistically, the expression of NADPH oxidase 1 (Nox1) was found to be suppressed and antioxidant enzymes including catalase, superoxide dismutase 2 (SOD-2), and heme oxygenase-1(HO-1) were enhanced following RH treatment, suggesting RH exhibited antioxidant activity by reducing the generation of reactive oxygen species (ROS) as well as enhancing the depletion of ROS. rhapontin 212-214 heme oxygenase 1 Homo sapiens 182-186 34638586-13 2021 The upregulation of NEDD4-1 attenuated PTEN expression and promoted the AKT/NRF2/HO-1 oxidative stress signaling axis, which in turn conferred amplified defense against reactive oxygen species (ROS) and eventually higher resistance against TMZ treatment. Reactive Oxygen Species 194-197 heme oxygenase 1 Homo sapiens 81-85 34638586-13 2021 The upregulation of NEDD4-1 attenuated PTEN expression and promoted the AKT/NRF2/HO-1 oxidative stress signaling axis, which in turn conferred amplified defense against reactive oxygen species (ROS) and eventually higher resistance against TMZ treatment. Temozolomide 240-243 heme oxygenase 1 Homo sapiens 81-85 34638586-15 2021 CONCLUSIONS: These findings demonstrate the critical role of NEDD4-1 in regulating the redox imbalance in TMZ-resistant GBM cells via the degradation of PTEN and the upregulation of the AKT/NRF2/HO-1 signaling pathway. Temozolomide 106-109 heme oxygenase 1 Homo sapiens 195-199 34214916-5 2021 Low doses of DMEP upregulated nuclear factor E2-related factor 2 (Nrf2) and downstream protein haeme oxygenase-1 (HO-1) levels and high doses down-regulated their levels. dimethoxyethyl phthalate 13-17 heme oxygenase 1 Homo sapiens 95-112 34680412-5 2021 The human apoptosis array and Western blot assay showed that magnolol increased the expression of cleaved caspase-3 proteins and heme oxygenase-1 (HO-1). magnolol 61-69 heme oxygenase 1 Homo sapiens 129-145 34680412-5 2021 The human apoptosis array and Western blot assay showed that magnolol increased the expression of cleaved caspase-3 proteins and heme oxygenase-1 (HO-1). magnolol 61-69 heme oxygenase 1 Homo sapiens 147-151 34214916-5 2021 Low doses of DMEP upregulated nuclear factor E2-related factor 2 (Nrf2) and downstream protein haeme oxygenase-1 (HO-1) levels and high doses down-regulated their levels. dimethoxyethyl phthalate 13-17 heme oxygenase 1 Homo sapiens 114-118 34455781-5 2021 Besides, the EPS-b by its scavenging potential also maintained the oxidative balance in the Caco-2 cells under oxidative stress and preserved the cellular antioxidative defense system (CAT, GPx, SOD, HO1, and GCLC) at the basal level. N-ethyl-3-piperidyl benzilate 13-18 heme oxygenase 1 Homo sapiens 200-203 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 heme oxygenase 1 Homo sapiens 206-210 34573068-6 2021 Moreover, BTDE could inhibit the expression of Kelch-like epichlorohydrin-associated protein 1 (Keap1) and increase the expression of both nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream proteins TrXR1, HO-1, and NQO1. btde 10-14 heme oxygenase 1 Homo sapiens 225-229 34566656-7 2021 In addition, JSH-23 attenuated H2O2-induced apoptosis and mineralization reduction in osteoblasts by reducing ROS production and enhancing Nrf2/HO-1 expression. Hydrogen Peroxide 31-35 heme oxygenase 1 Homo sapiens 144-148 34646329-12 2021 Molecular docking revealed a good binding affinity of active components (quercetin, bisdemethoxycurcumin, and kaempferol) with the corresponding targets (CYP3A4, CYP1A1, HMOX1, DRD2, DPP4, ADRA2A, ADRA2C, NR1I2, and LGALS4). Quercetin 73-82 heme oxygenase 1 Homo sapiens 170-175 34646329-12 2021 Molecular docking revealed a good binding affinity of active components (quercetin, bisdemethoxycurcumin, and kaempferol) with the corresponding targets (CYP3A4, CYP1A1, HMOX1, DRD2, DPP4, ADRA2A, ADRA2C, NR1I2, and LGALS4). bisdemethoxycurcumin 84-104 heme oxygenase 1 Homo sapiens 170-175 34646329-12 2021 Molecular docking revealed a good binding affinity of active components (quercetin, bisdemethoxycurcumin, and kaempferol) with the corresponding targets (CYP3A4, CYP1A1, HMOX1, DRD2, DPP4, ADRA2A, ADRA2C, NR1I2, and LGALS4). kaempferol 110-120 heme oxygenase 1 Homo sapiens 170-175 34575836-11 2021 The effects of compound 1 were partly reversed by co-treatment with a HO-1 inhibitor, tin protoporphyrin IX. tin protoporphyrin IX 86-107 heme oxygenase 1 Homo sapiens 70-74 34572050-3 2021 Subcellular traffics of HO-1 to different organelles constitute a network of interactions compromising a variety of effectors such as pro-oxidants, ROS, mitochondrial enzymes, and nucleic transcription factors. ros 148-151 heme oxygenase 1 Homo sapiens 24-28 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Bilirubin 74-83 heme oxygenase 1 Homo sapiens 29-33 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). epoxyeicosatrienoic acid 153-177 heme oxygenase 1 Homo sapiens 29-33 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). EET 179-182 heme oxygenase 1 Homo sapiens 29-33 34081904-8 2021 Therefore, this review summarizes the biological characteristics of HO-1, and the relationship between inflammatory response and reactive oxygen species production regulated by HO-1 and osteoporosis. Reactive Oxygen Species 129-152 heme oxygenase 1 Homo sapiens 68-72 34081904-8 2021 Therefore, this review summarizes the biological characteristics of HO-1, and the relationship between inflammatory response and reactive oxygen species production regulated by HO-1 and osteoporosis. Reactive Oxygen Species 129-152 heme oxygenase 1 Homo sapiens 177-181 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). omega-3 185-192 heme oxygenase 1 Homo sapiens 29-33 34118221-10 2021 This study suggests that ALA priming may improve the therapeutic potential of ADSCs against chronic liver problems by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant factors heme oxygenase 1 (HO-1) and quinone acceptor oxidoreductase-1 (NQO1). Thioctic Acid 25-28 heme oxygenase 1 Homo sapiens 223-239 34118221-10 2021 This study suggests that ALA priming may improve the therapeutic potential of ADSCs against chronic liver problems by activating the nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant factors heme oxygenase 1 (HO-1) and quinone acceptor oxidoreductase-1 (NQO1). Thioctic Acid 25-28 heme oxygenase 1 Homo sapiens 241-245 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Fatty Acids, Unsaturated 203-230 heme oxygenase 1 Homo sapiens 29-33 34499923-5 2022 Recent reports indicate that HO-1 with its antioxidants via the effect of bilirubin increases formation of biologically active lipid metabolites such as epoxyeicosatrienoic acid (EET), omega-3 and other polyunsaturated fatty acids (PUFAs). Fatty Acids, Unsaturated 232-237 heme oxygenase 1 Homo sapiens 29-33 34518768-8 2021 Further experiments showed that CR 8 : 2 exhibited significantly greater effects in increasing Nrf2 translocation and expressions of Nrf2 and HO-1 proteins, as well as SOD activity and total cellular NAD production, than that of C or R alone. Chromium 32-34 heme oxygenase 1 Homo sapiens 142-146 34365685-0 2021 Melatonin induces Nrf2-HO-1 reprogramming and corrections in hepatic core clock oscillations in Non-alcoholic fatty liver disease. Melatonin 0-9 heme oxygenase 1 Homo sapiens 23-27 34153850-6 2021 In addition, curcumin pretreatment significantly increased the expression of nuclear Nrf2, and the productions of superoxide dismutase 1 and heme oxygenase-1, which were the target anti-oxidative enzymes of the Keap1/Nrf2/ARE pathway. Curcumin 13-21 heme oxygenase 1 Homo sapiens 141-157 34306210-9 2021 In addition, salinomycin significantly downregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1, NAD(P)H quinone dehydrogenase 1 and glutamate-cysteine ligase catalytic subunit and decreased the activity of the antioxidant enzymes superoxide dismutase, catalase and glutathione peroxidase in PC-3 and DU145 cells. salinomycin 13-24 heme oxygenase 1 Homo sapiens 123-139 34314818-6 2021 EAA treatment increased nuclear Nrf2 translocation via the p38, PKC, and ROS pathways leading to HO-1, gamma-GCLC, and NQO-1 antioxidant expression in HaCaT cells. 3-O-ethylascorbic acid 0-3 heme oxygenase 1 Homo sapiens 97-101 34426902-10 2021 Moreover, the expression of HO-1 and NQO-1 was upregulated in hASCs pretreated with DMF which confirms the activation of the Nrf2 pathway. Dimethyl Fumarate 84-87 heme oxygenase 1 Homo sapiens 28-32 34296312-0 2021 Dimethyl itaconate inhibits LPS-induced microglia inflammation and inflammasome-mediated pyroptosis via inducing autophagy and regulating the Nrf-2/HO-1 signaling pathway. dimethyl itaconate 0-18 heme oxygenase 1 Homo sapiens 148-152 34296312-9 2021 In addition, DI effectively reduced reactive oxygen species production through the Nrf-2/HO-1 pathway. dimethyl itaconate 13-15 heme oxygenase 1 Homo sapiens 89-93 34296312-9 2021 In addition, DI effectively reduced reactive oxygen species production through the Nrf-2/HO-1 pathway. Reactive Oxygen Species 36-59 heme oxygenase 1 Homo sapiens 89-93 34197898-7 2021 We demonstrated that the drug exerted its cytoprotective effects by activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme-oxygenase (HO)-1 pathway, which in turn, is dependent on AKT activation by metformin. Metformin 210-219 heme oxygenase 1 Homo sapiens 130-151 34106394-11 2021 NaB also suppressed the BAX nuclear translocation and modulated Nrf-2, HO-1 and MnSOD expression in neuroblastoma cells. nab 0-3 heme oxygenase 1 Homo sapiens 71-75 34573035-0 2021 Bilirubin Links HO-1 and UGT1A1*28 Gene Polymorphisms to Predict Cardiovascular Outcome in Patients Receiving Maintenance Hemodialysis. Bilirubin 0-9 heme oxygenase 1 Homo sapiens 16-20 34425169-7 2021 While the progression of gene changes was different for each subject, a common pattern was observed in arsenic treated cells over time, with early upregulation of oxidative stress responses (HMOX1, NQ01, TXN, TXNRD1) and down-regulation of immune/inflammatory responses (IKKalpha). Arsenic 103-110 heme oxygenase 1 Homo sapiens 191-196 34216683-10 2021 The increase in nuclear Nrf2, HO-1 and NQO1 expression induced by isoflurane was amplified by treatment with BCA. Isoflurane 66-76 heme oxygenase 1 Homo sapiens 30-34 34573035-1 2021 Serum bilirubin levels, which are determined by a complex interplay of various enzymes, including heme oxygenase-1 (HO-1) and uridine diphosphate-glucuronosyl transferase (UGT1A1), may be protective against progression of cardiovascular disease (CVD) in hemodialysis patients. Bilirubin 6-15 heme oxygenase 1 Homo sapiens 98-114 34427071-12 2021 Our results also showed that temozolomide-induced ferroptosis is associated with regulation of the Nrf2/HO-1 pathway. Temozolomide 29-41 heme oxygenase 1 Homo sapiens 104-108 34573035-1 2021 Serum bilirubin levels, which are determined by a complex interplay of various enzymes, including heme oxygenase-1 (HO-1) and uridine diphosphate-glucuronosyl transferase (UGT1A1), may be protective against progression of cardiovascular disease (CVD) in hemodialysis patients. Bilirubin 6-15 heme oxygenase 1 Homo sapiens 116-120 34573035-3 2021 This retrospective study enrolled 1080 prevalent hemodialysis patients and the combined genetic polymorphisms of HO-1 and UGT1A1 on serum bilirubin were analyzed. Bilirubin 138-147 heme oxygenase 1 Homo sapiens 113-117 34573035-5 2021 Mean serum bilirubin was highest in patients with S/S + S/L of the HO-1 promoter and UGT1A1 7/7 genotypes (Group 1), intermediate in those with S/S + S/L of the HO-1 promoter and UGT1A1 7/6 + 6/6 genotypes (Group 2), and lowest in the carriers with the L/L HO-1 promoter and UGT1A1 7/6 + 6/6 genotypes (Group 3) (p < 0.001). Bilirubin 11-20 heme oxygenase 1 Homo sapiens 67-71 34573035-5 2021 Mean serum bilirubin was highest in patients with S/S + S/L of the HO-1 promoter and UGT1A1 7/7 genotypes (Group 1), intermediate in those with S/S + S/L of the HO-1 promoter and UGT1A1 7/6 + 6/6 genotypes (Group 2), and lowest in the carriers with the L/L HO-1 promoter and UGT1A1 7/6 + 6/6 genotypes (Group 3) (p < 0.001). Bilirubin 11-20 heme oxygenase 1 Homo sapiens 161-165 34504854-4 2021 Heme oxygenase-1 (HO-1), an enzyme involved in the process of heme degradation, has attracted widespread attention in recent years due to its cytoprotective properties. Heme 62-66 heme oxygenase 1 Homo sapiens 0-16 34503136-0 2021 Chrysosplenol D Triggers Apoptosis through Heme Oxygenase-1 and Mitogen-Activated Protein Kinase Signaling in Oral Squamous Cell Carcinoma. chrysosplenol D 0-15 heme oxygenase 1 Homo sapiens 43-59 34503136-7 2021 Moreover, the upregulation of heme oxygenase-1 (HO-1) was found to be critical for chrysosplenol D-induced apoptotic cell death. Chrysosplenol C 83-96 heme oxygenase 1 Homo sapiens 30-46 34503136-7 2021 Moreover, the upregulation of heme oxygenase-1 (HO-1) was found to be critical for chrysosplenol D-induced apoptotic cell death. Chrysosplenol C 83-96 heme oxygenase 1 Homo sapiens 48-52 34503136-9 2021 The findings of the present study indicated that chrysosplenol D exerts anticancer effects on OSCC by suppressing the MAPK pathway and activating HO-1 expression. chrysosplenol D 49-64 heme oxygenase 1 Homo sapiens 146-150 34217685-5 2021 Furthermore, it was demonstrated that luteolin promoted Nrf2 nuclear translocation, thereby increasing the expression of HO-1 that reduces ROS production, while the anti-pyroptotic effect of luteolin was reversed by a specific Nrf2 inhibitor. ros 139-142 heme oxygenase 1 Homo sapiens 121-125 34504854-4 2021 Heme oxygenase-1 (HO-1), an enzyme involved in the process of heme degradation, has attracted widespread attention in recent years due to its cytoprotective properties. Heme 62-66 heme oxygenase 1 Homo sapiens 18-22 34504854-7 2021 Many strategies, such as the administration of HO-1-overexpressing macrophages, use of phytochemicals, and carbon monoxide-based therapies, have been developed to target HO-1 in a variety of nephropathological animal models, indicating that HO-1 is a promising protein for the treatment of kidney diseases. Carbon Monoxide 107-122 heme oxygenase 1 Homo sapiens 170-174 34483918-0 2021 Nicardipine Inhibits Breast Cancer Migration via Nrf2/HO-1 Axis and Matrix Metalloproteinase-9 Regulation. Nicardipine 0-11 heme oxygenase 1 Homo sapiens 54-58 34557667-12 2021 Notably, the administration of dimethyl itaconate to compensate for the deficiency of immunoresponsive gene 1/itaconate axis led to enhanced microglial heme oxygenase-1 expression, alleviated ischaemic brain injury, improved motor function and decreased mortality in IRG1-/- stroke animals. dimethyl itaconate 31-49 heme oxygenase 1 Homo sapiens 152-168 34557667-12 2021 Notably, the administration of dimethyl itaconate to compensate for the deficiency of immunoresponsive gene 1/itaconate axis led to enhanced microglial heme oxygenase-1 expression, alleviated ischaemic brain injury, improved motor function and decreased mortality in IRG1-/- stroke animals. itaconic acid 110-119 heme oxygenase 1 Homo sapiens 152-168 34485154-8 2021 5-FU promoted the expression of Keap1 and increased the binding to NF-E2-related factor 2 (Nrf2) to reduce the nuclear translocation of Nrf2, thereby weakening the transcriptional activity of Nrf2 to inhibit the expression of HO-1; reducing the activity of GSH, SOD, and CAT to increase ROS content; and aggravating DNA damage (indicated by the increase in 8-OHdG). Fluorouracil 0-4 heme oxygenase 1 Homo sapiens 226-230 34602929-8 2021 In addition, the effects of TMF on Abeta25-35 toxicity were related to the upregulation of phase II antioxidant and nuclear factor erythroid 2-related factor-2 (Nrf2) signaling, including superoxide dismutase (SOD), heme oxygenase (HO)-1, and nuclear translocation of Nrf2. 5,6,7,4'-tetramethoxyflavanone 28-31 heme oxygenase 1 Homo sapiens 216-237 34445549-7 2021 In addition, western blot analysis also demonstrated that kaempferol and kaempferide reduced expression of heme oxygenase-1 (HO-1) and nuclear transcription factor-erythroid 2-related factor 2 (Nrf2). kaempferol 58-68 heme oxygenase 1 Homo sapiens 107-123 34445549-7 2021 In addition, western blot analysis also demonstrated that kaempferol and kaempferide reduced expression of heme oxygenase-1 (HO-1) and nuclear transcription factor-erythroid 2-related factor 2 (Nrf2). kaempferol 58-68 heme oxygenase 1 Homo sapiens 125-129 34445549-7 2021 In addition, western blot analysis also demonstrated that kaempferol and kaempferide reduced expression of heme oxygenase-1 (HO-1) and nuclear transcription factor-erythroid 2-related factor 2 (Nrf2). kaempferide 73-84 heme oxygenase 1 Homo sapiens 107-123 34445549-7 2021 In addition, western blot analysis also demonstrated that kaempferol and kaempferide reduced expression of heme oxygenase-1 (HO-1) and nuclear transcription factor-erythroid 2-related factor 2 (Nrf2). kaempferide 73-84 heme oxygenase 1 Homo sapiens 125-129 34087331-13 2021 RT-PCR confirmed the HMOX1 upregulation and ATG12 downregulation protected the PC-3rst23 cells from SRJ23 cytotoxicity. srj23 100-105 heme oxygenase 1 Homo sapiens 21-26 34483918-10 2021 The inhibition of HO-1 abrogates nicardipine-reduced matrix metalloproteinase-9 expression. Nicardipine 33-44 heme oxygenase 1 Homo sapiens 18-22 34483918-11 2021 Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. Carbon Monoxide 44-46 heme oxygenase 1 Homo sapiens 30-34 34483918-11 2021 Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. ammonium ferrous sulfate 48-52 heme oxygenase 1 Homo sapiens 30-34 34483918-11 2021 Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. Biliverdine 58-68 heme oxygenase 1 Homo sapiens 30-34 34483918-11 2021 Moreover, the end products of HO-1, namely, CO, Fe2+, and biliverdin (will converted to bilirubin), also decreases the expression of matrix metalloproteinase-9. Bilirubin 88-97 heme oxygenase 1 Homo sapiens 30-34 34483918-12 2021 Conclusion: These findings suggest that nicardipine-mediated matrix metalloproteinase-9 reduction is regulated by Nrf2/HO-1 axis and its catalytic end products. Nicardipine 40-51 heme oxygenase 1 Homo sapiens 119-123 34483918-9 2021 In addition, we also revealed that nicardipine increases the Nrf2 and HO-1 expression. Nicardipine 35-46 heme oxygenase 1 Homo sapiens 70-74 34369898-0 2022 Effect of Ginsenoside Rh1 on proliferation, apoptosis, and oxidative stress in vascular endothelial cells via regulation of the Nrf2/HO-1 signaling pathway. ginsenoside Rh1 10-25 heme oxygenase 1 Homo sapiens 133-137 34456904-8 2021 ME markedly increased the expression of Nrf2, thus resulting in a higher level of HO-1. methionylglutamic acid 0-2 heme oxygenase 1 Homo sapiens 82-86 34452191-5 2021 Hemin (HO-1 inducer) induced HO-1 mRNA and protein expression, as expected, and below 50 mM, dose-dependently reduced the viral RNA and proteins in the HAV-infected cells without cytotoxicity. Hemin 0-5 heme oxygenase 1 Homo sapiens 7-11 34452191-5 2021 Hemin (HO-1 inducer) induced HO-1 mRNA and protein expression, as expected, and below 50 mM, dose-dependently reduced the viral RNA and proteins in the HAV-infected cells without cytotoxicity. Hemin 0-5 heme oxygenase 1 Homo sapiens 29-33 34452191-7 2021 Although ZnPP-9, an HO-1 inhibitor, did not affect HAV replication, it significantly inhibited hemin-induced antiviral activity in HAV-infected cells. zinc protoporphyrin 9-15 heme oxygenase 1 Homo sapiens 20-24 34452191-8 2021 Additionally, FeCl3, CORM-3, biliverdin, and the HO-1 inducers andrographolide and CoPP inhibited HAV replication in the HAV-infected cells; andrographolide and CoPP exhibited a dose-dependent effect. andrographolide 63-78 heme oxygenase 1 Homo sapiens 49-53 34452191-8 2021 Additionally, FeCl3, CORM-3, biliverdin, and the HO-1 inducers andrographolide and CoPP inhibited HAV replication in the HAV-infected cells; andrographolide and CoPP exhibited a dose-dependent effect. COPP protocol 83-87 heme oxygenase 1 Homo sapiens 49-53 34369898-7 2022 Moreover, ginsenoside Rh1 also relieved oxidative stress in ox-LDL-treated VECs by activating the Nef2/HO-1 signaling pathway. ginsenoside Rh1 10-25 heme oxygenase 1 Homo sapiens 103-107 34369898-8 2022 Thus, ginsenoside Rh1 affects the proliferation, apoptosis, and oxidative stress in ox-LDL-treated VECs by activating the Nef2/HO-1 signaling pathway. ginsenoside Rh1 6-21 heme oxygenase 1 Homo sapiens 127-131 34313971-12 2021 Moreover, A-EVs significantly upregulated the mRNA expression of Nrf2, HO-1, CAT, and SOD genes in H2O2-treated HaCaT cells. a-evs 10-15 heme oxygenase 1 Homo sapiens 71-75 34361114-8 2021 The results of the present study demonstrate that pDCs can respond to LPS and that quercetin exposure modulates soluble factors release through the same molecular pathway used by mDCs (Slpi, Hmox1, and AP-1). Quercetin 83-92 heme oxygenase 1 Homo sapiens 191-196 34338265-6 2021 Further study showed that the protective effect of kaempferol was associated with an enhanced mRNA level of genes related to cell cycle progression (cyclin D1, CDK4, and E2F1) and genes implicated in the anti-oxidant system (GSR, GSTA4, and HO-1), up-regulated abundance of tight junctions (ZO-1, ZO-2, occludin, and claudin-4), as well as enhanced Nrf2, an anti-oxidant transcription factor. kaempferol 51-61 heme oxygenase 1 Homo sapiens 241-245 34382871-2 2021 In this study, the effects of CYJ-27 on the regulation of inducible nitric oxide synthase (iNOS), heme oxygenase (HO)-1, and cyclooxygenase (COX-)2 were characterized in lipopolysaccharide (LPS)-treated human umbilical vein endothelial cells (HUVECs). cyj-27 30-36 heme oxygenase 1 Homo sapiens 98-139 34175589-6 2021 RESULTS: Obesity-associated AHR is ameliorated by administration of BCP by inhibition of the macrophage polarization by activation of AMPKalpha, Nrf2/HO-1 and AdipoR1 and AdipoR2 signaling pathway, up-regulation of adiponectin, GLP-1, IFN-gamma, SOD, catalase and down-regulation of NF-kappaB, leptin, IL-4, TNF, and IL-1beta. caryophyllene 68-71 heme oxygenase 1 Homo sapiens 150-154 34313971-12 2021 Moreover, A-EVs significantly upregulated the mRNA expression of Nrf2, HO-1, CAT, and SOD genes in H2O2-treated HaCaT cells. Hydrogen Peroxide 99-103 heme oxygenase 1 Homo sapiens 71-75 34360702-5 2021 Overexpression of Nrf2 or its target gene heme oxygenase 1 (HO1) significantly blocked the ROS accumulation in MCF-7 cells under T-2 toxin treatment. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 42-58 34361019-6 2021 Here, we examined whether pharmacological induction of HO-1 by cobalt protoporphyrin (CoPP) ameliorates cholestatic liver injury. cobaltiprotoporphyrin 63-84 heme oxygenase 1 Homo sapiens 55-59 34361019-6 2021 Here, we examined whether pharmacological induction of HO-1 by cobalt protoporphyrin (CoPP) ameliorates cholestatic liver injury. cobaltiprotoporphyrin 86-90 heme oxygenase 1 Homo sapiens 55-59 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Heme 114-118 heme oxygenase 1 Homo sapiens 0-16 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Heme 114-118 heme oxygenase 1 Homo sapiens 18-22 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Carbon Monoxide 145-160 heme oxygenase 1 Homo sapiens 0-16 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Carbon Monoxide 145-160 heme oxygenase 1 Homo sapiens 18-22 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Iron 162-166 heme oxygenase 1 Homo sapiens 0-16 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Iron 162-166 heme oxygenase 1 Homo sapiens 18-22 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Biliverdine 172-182 heme oxygenase 1 Homo sapiens 0-16 34360940-3 2021 Heme oxygenase-1 (HO-1) has evolved to promptly attend to such injurious potential by facilitating degradation of heme into equimolar amounts of carbon monoxide, iron, and biliverdin. Biliverdine 172-182 heme oxygenase 1 Homo sapiens 18-22 34321570-2 2021 Quantifying circadian disruption by analyzing expression of the circadian gene period circadian regulator 2 (PER2) and heme oxygenase 1 (HO1), which determines heme turnover, may prove to be potential diagnostic tools. Heme 160-164 heme oxygenase 1 Homo sapiens 119-135 34321570-2 2021 Quantifying circadian disruption by analyzing expression of the circadian gene period circadian regulator 2 (PER2) and heme oxygenase 1 (HO1), which determines heme turnover, may prove to be potential diagnostic tools. Heme 160-164 heme oxygenase 1 Homo sapiens 137-140 34312792-12 2022 Associations of early low HO-1 expression with vasospasm and late elevated HO-1 expression with DCI may point to detrimental effects of late HO-1 induction, warranting the need for further investigation in a larger study population. dci 96-99 heme oxygenase 1 Homo sapiens 75-79 34360702-5 2021 Overexpression of Nrf2 or its target gene heme oxygenase 1 (HO1) significantly blocked the ROS accumulation in MCF-7 cells under T-2 toxin treatment. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 60-63 34290541-3 2021 In 2018, we reported that heme oxygenase-1 (HO-1), an inducible stress response protein important for heme catabolism and implicated in PD pathology, was higher in PD saliva relative to healthy controls, suggesting that salivary HO-1 may serve as a potential biomarker of PD. Heme 102-106 heme oxygenase 1 Homo sapiens 44-48 34305600-6 2021 At the same time, the GN combination could also inhibit the H2O2-induced inflammatory response and apoptosis of human umbilical vein endothelial cells (HUVECs), which is mainly related to the activation of the AMPK/mTOR pathway by GN combination, which in turn induces the activation of Nrf2/HO-1 signal. Hydrogen Peroxide 60-64 heme oxygenase 1 Homo sapiens 292-296 34439417-7 2021 The cellular components mediating the downregulation of ROS included extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and uncoupling protein 2 (UCP2). Reactive Oxygen Species 56-59 heme oxygenase 1 Homo sapiens 173-189 34439417-7 2021 The cellular components mediating the downregulation of ROS included extracellular signal-regulated kinase 1/2 (ERK1/2), nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and uncoupling protein 2 (UCP2). Reactive Oxygen Species 56-59 heme oxygenase 1 Homo sapiens 191-195 34264023-10 2022 Carcinoma cells with high HO-1 levels were significantly associated with pathological resistance to NACRT. nacrt 100-105 heme oxygenase 1 Homo sapiens 26-30 34264023-13 2022 HO-1 levels in lymph node metastases could be used to predict the eventual clinical outcome of patients with esophageal cancer receiving NACRT. nacrt 137-142 heme oxygenase 1 Homo sapiens 0-4 34290541-3 2021 In 2018, we reported that heme oxygenase-1 (HO-1), an inducible stress response protein important for heme catabolism and implicated in PD pathology, was higher in PD saliva relative to healthy controls, suggesting that salivary HO-1 may serve as a potential biomarker of PD. Heme 102-106 heme oxygenase 1 Homo sapiens 26-42 34228907-10 2021 Additionally, Isoeucommin A protected H2 O2 -stimulated renal tubular epithelial cells (RTECs) from oxidative stress and activated Nrf2/HO-1 signaling pathway in high glucose-stimulated human renal mesangial cells (HRMCs). Isoeucommin A 14-27 heme oxygenase 1 Homo sapiens 136-140 34228907-10 2021 Additionally, Isoeucommin A protected H2 O2 -stimulated renal tubular epithelial cells (RTECs) from oxidative stress and activated Nrf2/HO-1 signaling pathway in high glucose-stimulated human renal mesangial cells (HRMCs). Hydrogen Peroxide 38-43 heme oxygenase 1 Homo sapiens 136-140 34228907-11 2021 In conclusion, Isoeucommin A could alleviate inflammation and oxidative stress in in vitro and in vivo DN models and thus attenuate kidney injury by activating the Nrf2/HO-1 signaling pathway. Isoeucommin A 15-28 heme oxygenase 1 Homo sapiens 169-173 34209690-6 2021 Treatment with Hidrox was able to improve CYP-induced inflammation and oxidative stress via the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. hidrox 15-21 heme oxygenase 1 Homo sapiens 148-164 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. cumoox 18-24 heme oxygenase 1 Homo sapiens 181-185 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. zinc protoporphyrin 37-59 heme oxygenase 1 Homo sapiens 181-185 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. zinc protoporphyrin 61-63 heme oxygenase 1 Homo sapiens 181-185 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. Copper 72-74 heme oxygenase 1 Homo sapiens 181-185 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. ccmz 76-80 heme oxygenase 1 Homo sapiens 181-185 34075731-2 2021 Herein, PEGylated CuMoOx -coated and zinc protoporphyrin IX (ZP)-loaded Cu (CCMZ) NPs are designed to afford rapid degradation ability and augmented CDT efficacy through inhibiting HO-1 activity and depleting GSH. cdt 149-152 heme oxygenase 1 Homo sapiens 181-185 34075731-5 2021 Furthermore, the release of ZP from CCMZ NPs can inhibit HO-1 activity and provide a favorable microenvironment for CDT, and the release of Cu and Mo ions can convert hydrogen peroxide into hydroxyl radical to eliminate tumor cells more efficiently. ccmz 36-40 heme oxygenase 1 Homo sapiens 57-61 34082940-6 2021 Compared with free BA, Exo-BA significantly reduced the generation of reactive oxygen species (ROS) and activated the Nrf2/HO-1 pathway in neurons, thus significantly alleviating cerebral ischemic injury in a stroke model. exo-ba 23-29 heme oxygenase 1 Homo sapiens 123-127 34209690-6 2021 Treatment with Hidrox was able to improve CYP-induced inflammation and oxidative stress via the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. hidrox 15-21 heme oxygenase 1 Homo sapiens 166-170 34345202-0 2021 Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. tagitinin 0-11 heme oxygenase 1 Homo sapiens 50-54 34345202-11 2021 As a downstream gene (effector) of Nrf2, heme oxygenase-1 (HO-1) expression increased significantly with the treatment of tagitinin C. Upregulated HO-1 led to the increase in the labile iron pool, which promoted lipid peroxidation, meanwhile tagitinin C showed synergistic anti-tumor effect together with erastin. Iron 186-190 heme oxygenase 1 Homo sapiens 147-151 34345202-13 2021 Mechanistically, tagitinin C induces ferroptosis through ER stress-mediated activation of PERK-Nrf2-HO-1 signaling pathway. tagitinin 17-28 heme oxygenase 1 Homo sapiens 100-104 34208670-5 2021 After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Hemin 42-47 heme oxygenase 1 Homo sapiens 6-10 34202670-0 2021 Cytoprotection against Oxidative Stress by Methylnissolin-3-O-beta-d-glucopyranoside from Astragalus membranaceus Mainly via the Activation of the Nrf2/HO-1 Pathway. methylnissolin-3-o-beta-d-glucopyranoside 43-84 heme oxygenase 1 Homo sapiens 152-156 34202670-5 2021 MNG induced the expression of Nrf2, HO-1 and NQO1, accelerated the translocation of Nrf2 into nuclei, and enhanced the phosphorylation of AKT. 1-Methyl-3-nitro-1-nitrosoguanidine 0-3 heme oxygenase 1 Homo sapiens 36-40 34202670-6 2021 The MNG-induced expression of Nrf2, HO-1, and NQO1 were abolished by Nrf2 siRNA, while the MNG-induced expression of Nrf2 and HO-1 was abated and the AKT phosphorylation was blocked by LY294002 (a PI3K inhibitor). 1-Methyl-3-nitro-1-nitrosoguanidine 4-7 heme oxygenase 1 Homo sapiens 36-40 34202670-6 2021 The MNG-induced expression of Nrf2, HO-1, and NQO1 were abolished by Nrf2 siRNA, while the MNG-induced expression of Nrf2 and HO-1 was abated and the AKT phosphorylation was blocked by LY294002 (a PI3K inhibitor). 1-Methyl-3-nitro-1-nitrosoguanidine 4-7 heme oxygenase 1 Homo sapiens 126-130 34202670-6 2021 The MNG-induced expression of Nrf2, HO-1, and NQO1 were abolished by Nrf2 siRNA, while the MNG-induced expression of Nrf2 and HO-1 was abated and the AKT phosphorylation was blocked by LY294002 (a PI3K inhibitor). 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 185-193 heme oxygenase 1 Homo sapiens 36-40 34202670-6 2021 The MNG-induced expression of Nrf2, HO-1, and NQO1 were abolished by Nrf2 siRNA, while the MNG-induced expression of Nrf2 and HO-1 was abated and the AKT phosphorylation was blocked by LY294002 (a PI3K inhibitor). 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 185-193 heme oxygenase 1 Homo sapiens 126-130 34202670-9 2021 The enhancement of Nrf2 and HO-1 by MNG upon H2O2 injury was reduced by LY294002. 1-Methyl-3-nitro-1-nitrosoguanidine 36-39 heme oxygenase 1 Homo sapiens 28-32 34202670-9 2021 The enhancement of Nrf2 and HO-1 by MNG upon H2O2 injury was reduced by LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 72-80 heme oxygenase 1 Homo sapiens 28-32 34202670-10 2021 These data showed that MNG protected EA.hy926 cells against oxidative damage through the Nrf2/HO-1 and at least partially the PI3K/Akt pathways. 1-Methyl-3-nitro-1-nitrosoguanidine 23-26 heme oxygenase 1 Homo sapiens 94-98 34135317-9 2021 Functionally, overexpression of HO-1 counteracted the promotional effects of SIRT4 on ROS accumulation and apoptosis. Reactive Oxygen Species 86-89 heme oxygenase 1 Homo sapiens 32-36 34135317-11 2021 By contrast, downregulation of p38-MAPK by SB203580 decreased intracellular ROS level and enhanced the expression of HO-1. SB 203580 43-51 heme oxygenase 1 Homo sapiens 117-121 34208670-5 2021 After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Glucose 142-149 heme oxygenase 1 Homo sapiens 6-10 34208670-5 2021 After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Adenosine Triphosphate 158-161 heme oxygenase 1 Homo sapiens 6-10 34208670-5 2021 After HO-1 pharmacological induction with hemin, PC3 and C4-2B cells exhibited a significantly impaired cellular metabolic rate, reflected by glucose uptake, ATP production, lactate dehydrogenase (LDH) activity and extracellular lactate levels. Lactic Acid 229-236 heme oxygenase 1 Homo sapiens 6-10 34306342-8 2021 UTI blocked ferroptosis-induced lipid peroxide accumulation by promoting nuclear translocation of NRF2 to activate its downstream targets (HO-1). Lipid Peroxides 32-46 heme oxygenase 1 Homo sapiens 139-143 34646076-14 2021 Conclusions: We concluded that d-Leu protects oocytes from psychological stress through the induction of HO-1 and SOD2 expression then by reducing oxidative stress. D-LEUCINE 31-36 heme oxygenase 1 Homo sapiens 105-109 34127699-5 2021 Furthermore, selumetinib reduced acrolein-induced increases in heme oxygenase-1 (a redox-regulated chaperone protein) and its transcriptional factor, Nrf-2 as well as FDP-lysine (acrolein-lysine adducts) and alpha-synuclein aggregation (a pathological biomarker of neurodegeneration). AZD 6244 13-24 heme oxygenase 1 Homo sapiens 63-79 34127699-8 2021 In conclusion, our study showed that selumetinib inhibited acrolein-activated Nrf-2-HO-1 pathway, acrolein-induced protein conjugation and aggregation as well as damage in neurite outgrowth and cell death, suggesting that selumetinib, a MEK-ERK inhibitor, may be a potential neuroprotective agent against acrolein-induced neurotoxicity in the CNS neurodegenerative diseases. AZD 6244 37-48 heme oxygenase 1 Homo sapiens 84-88 34127699-5 2021 Furthermore, selumetinib reduced acrolein-induced increases in heme oxygenase-1 (a redox-regulated chaperone protein) and its transcriptional factor, Nrf-2 as well as FDP-lysine (acrolein-lysine adducts) and alpha-synuclein aggregation (a pathological biomarker of neurodegeneration). Acrolein 33-41 heme oxygenase 1 Homo sapiens 63-79 34127699-8 2021 In conclusion, our study showed that selumetinib inhibited acrolein-activated Nrf-2-HO-1 pathway, acrolein-induced protein conjugation and aggregation as well as damage in neurite outgrowth and cell death, suggesting that selumetinib, a MEK-ERK inhibitor, may be a potential neuroprotective agent against acrolein-induced neurotoxicity in the CNS neurodegenerative diseases. Acrolein 59-67 heme oxygenase 1 Homo sapiens 84-88 34212035-3 2021 When treated with Carvacrol or tert-butylhydroquinone (TBHQ) that activates Nrf2, the expression of Keap1/Nrf2/HO-1, epithelial-mesenchymal transition- (EMT-) related proteins, and NALP3 was examined in OSCC cells. carvacrol 18-27 heme oxygenase 1 Homo sapiens 111-115 34208202-9 2021 Antioxidant properties of santamarine were also shown to arise from stimulating Nrf2-dependent expression of antioxidant enzymes SOD-1 and HO-1 in UVA-irradiated HDFs. santamarine 26-37 heme oxygenase 1 Homo sapiens 139-143 34212035-8 2021 Carvacrol significantly suppressed Keap1/Nrf2/HO-1 activation. carvacrol 0-9 heme oxygenase 1 Homo sapiens 46-50 34212035-9 2021 Carvacrol or silencing Nrf2 markedly inhibited the expression of Keap1/Nrf2/HO-1, EMT-related proteins, and NALP3 inflammasome in OSCC cells. carvacrol 0-9 heme oxygenase 1 Homo sapiens 76-80 34198827-7 2021 In addition, carnosic acid increased the translation of heme oxygenase-1 (HO-1), gamma-glutamylcysteine synthetase (gamma-GCSc), and glutathione S-transferase (GST) and both the translation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). salvin 13-26 heme oxygenase 1 Homo sapiens 56-72 34198827-7 2021 In addition, carnosic acid increased the translation of heme oxygenase-1 (HO-1), gamma-glutamylcysteine synthetase (gamma-GCSc), and glutathione S-transferase (GST) and both the translation and nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). salvin 13-26 heme oxygenase 1 Homo sapiens 74-78 34198827-8 2021 Taken together, these results indicate that carnosic acid could down-regulate ROS level in an early stage of MPI-induced adipocyte differentiation by attenuating ROS generation through suppression of NF-kappaB-mediated translation of Nox4 enzyme and increasing ROS neutralization through induction of Nrf2-mediated translation of phase II antioxidant enzymes such as HO-1, gamma-GCS, and GST, leading to its anti-adipogenetic effect. salvin 44-57 heme oxygenase 1 Homo sapiens 367-371 34198827-8 2021 Taken together, these results indicate that carnosic acid could down-regulate ROS level in an early stage of MPI-induced adipocyte differentiation by attenuating ROS generation through suppression of NF-kappaB-mediated translation of Nox4 enzyme and increasing ROS neutralization through induction of Nrf2-mediated translation of phase II antioxidant enzymes such as HO-1, gamma-GCS, and GST, leading to its anti-adipogenetic effect. Reactive Oxygen Species 78-81 heme oxygenase 1 Homo sapiens 367-371 34077680-7 2021 In addition, ZMO treatment markedly upregulated the expression of Nrf2 as well as its target genes, HO-1 and NAD(P)H:quinone oxidoreductase 1 in HepG2 cells. S-[2-({n-[(2s)-2-Hydroxy-3,3-Dimethyl-4-(Phosphonooxy)butanoyl]-Beta-Alanyl}amino)ethyl] (9z)-Hexadec-9-Enethioate 13-16 heme oxygenase 1 Homo sapiens 100-104 34199777-3 2021 Using an in vitro model, we demonstrated that HO-1 overexpression regulates stemness and resistance to 5-FU treatment, regardless of p53. Fluorouracil 103-107 heme oxygenase 1 Homo sapiens 46-50 34199777-5 2021 Carbon monoxide (CO) activated the ECE-1/endothelin-1 (ET-1) pathway, which could account for the protumoral effects of HO-1 in p53 wild-type cells, as demonstrated after treatment with bosentan (an antagonist of both ETRA and ETRB endothelin-1 receptors). Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 120-124 34199777-5 2021 Carbon monoxide (CO) activated the ECE-1/endothelin-1 (ET-1) pathway, which could account for the protumoral effects of HO-1 in p53 wild-type cells, as demonstrated after treatment with bosentan (an antagonist of both ETRA and ETRB endothelin-1 receptors). Bosentan 186-194 heme oxygenase 1 Homo sapiens 120-124 34199777-7 2021 In these cells, HO-1 could activate or inactivate certain unknown routes that could induce these contrary responses after treatment with bosentan in our cell model. Bosentan 137-145 heme oxygenase 1 Homo sapiens 16-20 35561750-9 2022 CoCl2-induced oxidative stress results in activation of the NRF2/HO-1-mediated cell survival pathway and inhibition of DNA repair, both of which were prevented by MSM. cobaltous chloride 0-5 heme oxygenase 1 Homo sapiens 65-69 34073678-7 2021 Heme degradation by heme oxygenase-1 (HO-1) is linked to cytoprotection via heme removal, as well as by activity-dependent end-product generation (i.e., bile pigments and CO), and other potential mechanisms. Heme 0-4 heme oxygenase 1 Homo sapiens 20-36 34073678-7 2021 Heme degradation by heme oxygenase-1 (HO-1) is linked to cytoprotection via heme removal, as well as by activity-dependent end-product generation (i.e., bile pigments and CO), and other potential mechanisms. Heme 0-4 heme oxygenase 1 Homo sapiens 38-42 34073678-7 2021 Heme degradation by heme oxygenase-1 (HO-1) is linked to cytoprotection via heme removal, as well as by activity-dependent end-product generation (i.e., bile pigments and CO), and other potential mechanisms. Heme 76-80 heme oxygenase 1 Homo sapiens 20-36 34073678-7 2021 Heme degradation by heme oxygenase-1 (HO-1) is linked to cytoprotection via heme removal, as well as by activity-dependent end-product generation (i.e., bile pigments and CO), and other potential mechanisms. Heme 76-80 heme oxygenase 1 Homo sapiens 38-42 34073678-8 2021 Therapeutic strategies targeting the heme/HO-1 pathway, including therapeutic modulation of heme levels, elevation (or inhibition) of HO-1 protein and activity, and application of CO donor compounds or gas show potential in inflammatory conditions including sepsis and pulmonary diseases. Heme 37-41 heme oxygenase 1 Homo sapiens 42-46 34065300-2 2021 In recent years, 5-ALA has been shown to possess anti-inflammatory and immunoregulatory properties through upregulation of heme oxygenase-1 via enhancement of porphyrin, indicating that it may be beneficial for the treatment of inflammatory conditions. 5-amino levulinic acid 17-22 heme oxygenase 1 Homo sapiens 123-139 34065584-7 2021 Form a molecular point of view, hydroxytyrosol is able to preserve the cellular redox balance and protein homeostasis by activating the Nrf2 pathway and increasing the expression of phase II detoxifying enzymes such as HO-1, SOD, Catalase, and GSH, thus counteracting the neurodegenerative damage. 3,4-dihydroxyphenylethanol 32-46 heme oxygenase 1 Homo sapiens 219-223 34104310-8 2021 In particular, the methanol extract was the most effective in protecting neuron-like SH-SY5Y cells against H2O2-induced oxidative stress by upregulating endogenous antioxidant enzymes such as thioredoxin reductase, heme oxygenase 1, NADPH quinone oxidoreductase, and glutathione reductase. Methanol 19-27 heme oxygenase 1 Homo sapiens 215-231 34104310-8 2021 In particular, the methanol extract was the most effective in protecting neuron-like SH-SY5Y cells against H2O2-induced oxidative stress by upregulating endogenous antioxidant enzymes such as thioredoxin reductase, heme oxygenase 1, NADPH quinone oxidoreductase, and glutathione reductase. Hydrogen Peroxide 107-111 heme oxygenase 1 Homo sapiens 215-231 34066673-6 2021 The expression of genes coding for antioxidant molecules, namely, heme oxygenase-1, alpha glutathione s-transferase, NF-kB, and liver fatty acid binding protein, were decreased due to benomyl. Benomyl 184-191 heme oxygenase 1 Homo sapiens 66-82 34277866-12 2021 Results: Photobiomodulation at 1, 2, and 3 J/cm2 combined with metformin significantly promoted diabetic cell lines of HPDLSCs viability (in MTT assay and ELISA reader of ROS, TNF-alpha, IL-10 results) and gene expression of Nrf2, Keap1, PIK3, and HO-1 levels (p< 0.05). Metformin 63-72 heme oxygenase 1 Homo sapiens 248-252 34121971-0 2021 Vinpocetine-based therapy is an attractive strategy against oxidative stress-induced hepatotoxicity in vitro by targeting Nrf2/HO-1 pathway. vinpocetine 0-11 heme oxygenase 1 Homo sapiens 127-131 34113558-12 2021 HMOX1 (up-regulated) and NME1-NME2 (down-regulated) were the main nodes in a PPI network consisting of 16 DEGs that were modulated by sporamin in the presence of TG. Thapsigargin 162-164 heme oxygenase 1 Homo sapiens 0-5 34069696-3 2021 In H2O2-induced Jukat T cells, haplopine (25 and 50 muM) suppressed the productions of proinflammatory cytokines (IL-4, IL-13, and COX-2) and increased the mRNA and protein expressions of oxidative stress defense enzymes (SOD, CAT, and HO-1) in a concentration-dependent manner. 4,8-dimethoxy-7-hydroxyfuro(2,3-b)quinoline 31-40 heme oxygenase 1 Homo sapiens 236-240 34064340-6 2021 The results showed that DHMBA protected mitochondrial function mainly during cold preservation, and suppressed cell death after rewarming, as shown by the MTT, ATP, mitochondrial membrane potential, LDH, and lipid peroxidation assays, together with enhanced survival signals (PI3K, Akt, p70S6K) and induction of antioxidant proteins (HO-1, NQO-1, TRX-1). 3,5-dihydroxy-4-methoxybenzyl alcohol 24-29 heme oxygenase 1 Homo sapiens 334-338 34277866-0 2021 Efficacy of Photobiomodulation and Metformin on Diabetic Cell Line of Human Periodontal Ligament Stem Cells through Keap1/Nrf2/Ho-1 Pathway. Metformin 35-44 heme oxygenase 1 Homo sapiens 127-131 34560924-9 2021 Our observations are the first to show that cardiac arrest followed by successful cardiopulmonary resuscitation results in significant alteration in cerebral concentrations of HO-1 and HO-2 levels indicating a prominent role of CO in brain pathology and methylene blue during CPR followed by induced hypothermia leading to superior neuroprotection after return of spontaneous circulation (ROSC), not reported earlier. Methylene Blue 254-268 heme oxygenase 1 Homo sapiens 176-180 35247778-14 2022 Further mechanistic study demonstrated that 7-d-GDN induced heme oxygenase-1 (HO-1), NAD(P)H dehydrogenase quinone 1(NQO1), which all participated in suppressing of oxidative stress. 7-d-gdn 44-51 heme oxygenase 1 Homo sapiens 60-76 35609387-17 2022 In addition, AU appears to reduce oxidative stress by upregulating NRF2/HO-1. aucubin 13-15 heme oxygenase 1 Homo sapiens 72-76 35618033-11 2022 Nevertheless, unlike HCQ, CQ is more likely to exaggerate intracellular prooxidant processes in activated starved microglia, which are inefficiently buffered by Nrf2/HO-1 signaling pathway activation. Chloroquine 26-28 heme oxygenase 1 Homo sapiens 166-170 35460631-1 2022 Carbon monoxide (CO), a member of the multifunctional gasotransmitters family produced by heme oxygenases (i.e., HO-1 and HO-2), has received significant attention because of its involvement in carbohydrate metabolism. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 113-117 35460631-1 2022 Carbon monoxide (CO), a member of the multifunctional gasotransmitters family produced by heme oxygenases (i.e., HO-1 and HO-2), has received significant attention because of its involvement in carbohydrate metabolism. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 113-117 35460631-1 2022 Carbon monoxide (CO), a member of the multifunctional gasotransmitters family produced by heme oxygenases (i.e., HO-1 and HO-2), has received significant attention because of its involvement in carbohydrate metabolism. Carbohydrates 194-206 heme oxygenase 1 Homo sapiens 113-117 35489465-7 2022 Our results showed that ZEA reduced the antioxidant capacity of the ST cells, induced the cell apoptosis and inhibited the gene and protein expression of Nrf2 and its downstream genes (ho-1,nqo1), while PC improved the cell antioxidant capacity, reduced the degree of ZEA-induced cell apoptosis and promoted the gene and protein expression of Nrf2 and its downstream genes. Zearalenone 24-27 heme oxygenase 1 Homo sapiens 185-189 35354089-0 2022 Cucurbitacin E glucoside alleviates concanavalin A-induced hepatitis through enhancing SIRT1/Nrf2/HO-1 and inhibiting NF-kB/NLRP3 signaling pathways. cucurbitacin E 0-14 heme oxygenase 1 Homo sapiens 98-102 35354089-0 2022 Cucurbitacin E glucoside alleviates concanavalin A-induced hepatitis through enhancing SIRT1/Nrf2/HO-1 and inhibiting NF-kB/NLRP3 signaling pathways. Glucosides 15-24 heme oxygenase 1 Homo sapiens 98-102 35452771-2 2022 Increasing lines of evidence show that T-2 toxin can reduce the levels of tight junction proteins, and nuclear factor erythroid 2-related factor 2 (Nrf2) by disrupting cellular barriers and the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) and Nrf2/heme oxygenase (HO)-1 pathways. Adenosine 201-210 heme oxygenase 1 Homo sapiens 259-285 35452771-2 2022 Increasing lines of evidence show that T-2 toxin can reduce the levels of tight junction proteins, and nuclear factor erythroid 2-related factor 2 (Nrf2) by disrupting cellular barriers and the cyclic adenosine monophosphate (cAMP)-protein kinase A (PKA) and Nrf2/heme oxygenase (HO)-1 pathways. Cyclic AMP 226-230 heme oxygenase 1 Homo sapiens 259-285 35500767-6 2022 Fucoidan targets multiple signaling systems, including Nrf2/HO-1, NF-kappaB, ERK and p38 MAPK, TGF-beta1, SIRT1, and GLP-1R signaling that are known to be associated with CKD pathobiology. fucoidan 0-8 heme oxygenase 1 Homo sapiens 60-64 35247778-14 2022 Further mechanistic study demonstrated that 7-d-GDN induced heme oxygenase-1 (HO-1), NAD(P)H dehydrogenase quinone 1(NQO1), which all participated in suppressing of oxidative stress. 7-d-gdn 44-51 heme oxygenase 1 Homo sapiens 78-82 35609009-3 2022 CGA antioxidant effects mediated through the Nrf2-heme oxygenase-1 signaling pathway were shown to enhance the levels of antioxidant enzymes such as superoxide dismutase, catalase, glutathione-S-transferases, glutathione peroxidase, and glutathione reductase as well as glutathione content. Superoxides 149-159 heme oxygenase 1 Homo sapiens 50-66 35475506-0 2022 3,4-Dihydroxyacetophenone attenuates oxidative stress-induced damage to HUVECs via regulation of the Nrf2/HO-1 pathway. 3,4-dihydroxyacetophenone 0-25 heme oxygenase 1 Homo sapiens 106-110 35475506-12 2022 In addition, high glucose upregulated Nrf2 and HO-1 protein and mRNA expression levels. Glucose 18-25 heme oxygenase 1 Homo sapiens 47-51 35475506-17 2022 3,4-DHAP markedly decreased the production of ROS, increased Nrf2 and HO-1 protein and mRNA expression levels, and promoted nuclear translocation of Nrf2 in HUVECs. 3,4-dihydroxyacetophenone 0-8 heme oxygenase 1 Homo sapiens 70-74 35475506-19 2022 Moreover, it was determined that compared to the 3,4-DHAP group, treatment with 3,4-DHAP and ML385 enhanced cell viability, and decreased ROS production, Nrf2 and HO-1 protein and mRNA expression levels, nuclear translocation of Nrf2, and LC3-II/LC3-I and PARP-1 protein expression levels. 3,4-dihydroxyacetophenone 49-57 heme oxygenase 1 Homo sapiens 163-167 35475506-19 2022 Moreover, it was determined that compared to the 3,4-DHAP group, treatment with 3,4-DHAP and ML385 enhanced cell viability, and decreased ROS production, Nrf2 and HO-1 protein and mRNA expression levels, nuclear translocation of Nrf2, and LC3-II/LC3-I and PARP-1 protein expression levels. 3,4-dihydroxyacetophenone 80-88 heme oxygenase 1 Homo sapiens 163-167 35475506-19 2022 Moreover, it was determined that compared to the 3,4-DHAP group, treatment with 3,4-DHAP and ML385 enhanced cell viability, and decreased ROS production, Nrf2 and HO-1 protein and mRNA expression levels, nuclear translocation of Nrf2, and LC3-II/LC3-I and PARP-1 protein expression levels. ML385 93-98 heme oxygenase 1 Homo sapiens 163-167 35475506-20 2022 Collectively, the results of the present study showed that 3,4-DHAP protected HUVECs against oxidative stress via regulation of the Nrf2/HO-1 pathway, by increasing autophagy and promoting DNA damage repair. 3,4-dihydroxyacetophenone 59-67 heme oxygenase 1 Homo sapiens 137-141 35635297-4 2022 In addition, LT significantly affected mitochondrial biogenesis and function and antioxidative related genes expression, and increased the protein expression of p-adenosine monophosphate (AMP)-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), nuclear factor E2-related factor 2 (Nrf2), NADPH quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) and decreased the Keap1 protein levels. Adenosine 163-172 heme oxygenase 1 Homo sapiens 396-412 35635297-4 2022 In addition, LT significantly affected mitochondrial biogenesis and function and antioxidative related genes expression, and increased the protein expression of p-adenosine monophosphate (AMP)-activated protein kinase (AMPK) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC-1alpha), nuclear factor E2-related factor 2 (Nrf2), NADPH quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) and decreased the Keap1 protein levels. Adenosine 163-172 heme oxygenase 1 Homo sapiens 414-418 35609009-3 2022 CGA antioxidant effects mediated through the Nrf2-heme oxygenase-1 signaling pathway were shown to enhance the levels of antioxidant enzymes such as superoxide dismutase, catalase, glutathione-S-transferases, glutathione peroxidase, and glutathione reductase as well as glutathione content. Glutathione 181-192 heme oxygenase 1 Homo sapiens 50-66 35609009-3 2022 CGA antioxidant effects mediated through the Nrf2-heme oxygenase-1 signaling pathway were shown to enhance the levels of antioxidant enzymes such as superoxide dismutase, catalase, glutathione-S-transferases, glutathione peroxidase, and glutathione reductase as well as glutathione content. Glutathione 209-220 heme oxygenase 1 Homo sapiens 50-66 35537139-11 2022 Meanwhile, the increased expression of SIRT1/PPAR-alpha and Nrf2/HO-1 pathways under the pretreatment of CPs in LO2 cells indicated that CPs could markedly relieve high fat-induced fatty liver injury, regulate insulin sensitivity, and reduce production of ROS. cps 105-108 heme oxygenase 1 Homo sapiens 65-69 35537139-11 2022 Meanwhile, the increased expression of SIRT1/PPAR-alpha and Nrf2/HO-1 pathways under the pretreatment of CPs in LO2 cells indicated that CPs could markedly relieve high fat-induced fatty liver injury, regulate insulin sensitivity, and reduce production of ROS. cps 137-140 heme oxygenase 1 Homo sapiens 65-69 35537139-11 2022 Meanwhile, the increased expression of SIRT1/PPAR-alpha and Nrf2/HO-1 pathways under the pretreatment of CPs in LO2 cells indicated that CPs could markedly relieve high fat-induced fatty liver injury, regulate insulin sensitivity, and reduce production of ROS. Reactive Oxygen Species 256-259 heme oxygenase 1 Homo sapiens 65-69 35598199-4 2022 METHODS AND RESULTS: Proteomic analyses found that ATO can affect the signaling pathway associated with ferroptosis, including the upregulation of iron absorption (FTL, FTH1, HO-1), ferritinophagy (LC3, P62, ATG7, NCOA4) and modifier of glutathione synthesis (GCLM); downregulation of glutamine synthetase (GS) and GPX4, which was the critical inhibitor of ferroptosis. Iron 147-151 heme oxygenase 1 Homo sapiens 175-179 35598290-0 2022 Hemin mitigates contrast-induced nephropathy by inhibiting ferroptosis via HO-1/Nrf2/GPX4 pathway. Hemin 0-5 heme oxygenase 1 Homo sapiens 75-79 35598290-9 2022 Furthermore, hemin"s induction of GPX4 involved HO-1 and nuclear factor Nrf2. Hemin 13-18 heme oxygenase 1 Homo sapiens 48-52 35598290-10 2022 Either HO-1 or Nrf2 inhibitor prevented hemin"s effect on GPX4 to a comparable extent, and over-expression of Nrf2 increased GPX4 expression. Hemin 40-45 heme oxygenase 1 Homo sapiens 7-11 35603834-7 2022 Fe-R-H regulators such as lactoferrin (LF), hemoxygenase-1 (HO-1), erythropoietin (EPO) and hepcidin modulators are innate bio-replenishments that sequester iron, neutralize iron-mediated free radicals, reduce oxidative stress, and improve host defense by optimizing iron metabolism. Iron 157-161 heme oxygenase 1 Homo sapiens 60-64 35603834-7 2022 Fe-R-H regulators such as lactoferrin (LF), hemoxygenase-1 (HO-1), erythropoietin (EPO) and hepcidin modulators are innate bio-replenishments that sequester iron, neutralize iron-mediated free radicals, reduce oxidative stress, and improve host defense by optimizing iron metabolism. Iron 174-178 heme oxygenase 1 Homo sapiens 60-64 35603834-7 2022 Fe-R-H regulators such as lactoferrin (LF), hemoxygenase-1 (HO-1), erythropoietin (EPO) and hepcidin modulators are innate bio-replenishments that sequester iron, neutralize iron-mediated free radicals, reduce oxidative stress, and improve host defense by optimizing iron metabolism. Iron 267-271 heme oxygenase 1 Homo sapiens 60-64 35603834-9 2022 Ferroptosis inhibitors such as ferrostatin-1, liproxstatin-1, quercetin, and melatonin could prevent mitochondrial lipid peroxidation, up-regulate antioxidant/GSH levels and abrogate iron overload-induced apoptosis through activation of Nrf2 and HO-1 signaling pathways. Quercetin 62-71 heme oxygenase 1 Homo sapiens 246-250 35598290-12 2022 Thus, we showed hemin mitigated CIN, inhibiting oxidative stress and ferroptosis, by upregulation of GPX4 via activation of HO-1/Nrf2. Hemin 16-21 heme oxygenase 1 Homo sapiens 124-128 35603834-9 2022 Ferroptosis inhibitors such as ferrostatin-1, liproxstatin-1, quercetin, and melatonin could prevent mitochondrial lipid peroxidation, up-regulate antioxidant/GSH levels and abrogate iron overload-induced apoptosis through activation of Nrf2 and HO-1 signaling pathways. Melatonin 77-86 heme oxygenase 1 Homo sapiens 246-250 35472411-3 2022 There is a significant body of research investigating the effects of oxidative stress/ROS on ASM behaviour, falling into the following categories; cigarette smoke and associated compounds, air pollutants, aero-allergens, asthma and COPD relevant mediators, and the anti-oxidant Nrf2/HO-1 signalling pathway. ros 86-89 heme oxygenase 1 Homo sapiens 283-287 35584686-7 2022 RESULTS: mRNA expression of HO-1, GCLC, HSPB1, VEGFA, PDGFA and PDGFB were significantly increased in ARPE-19 cells treated with H2O2 + hUCESC-CM compared to cells treated with H2O2 only. Hydrogen Peroxide 129-133 heme oxygenase 1 Homo sapiens 28-32 35605918-12 2022 The mechanism underlying the effect of DPH5 in alleviating IR was related to the PI3K/AKT- and Nrf2/HO-1-mediated regulation of the GSK3beta signaling pathway, and the results were further confirmed using the specific inhibitors LY294002 and ML385. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 229-237 heme oxygenase 1 Homo sapiens 100-104 35605918-12 2022 The mechanism underlying the effect of DPH5 in alleviating IR was related to the PI3K/AKT- and Nrf2/HO-1-mediated regulation of the GSK3beta signaling pathway, and the results were further confirmed using the specific inhibitors LY294002 and ML385. ML385 242-247 heme oxygenase 1 Homo sapiens 100-104 35620405-0 2022 11,12-Diacetyl-carnosol Protects SH-SY5Y Cells from Hydrogen Peroxide Damage through the Nrf2/HO-1 Pathway. Hydrogen Peroxide 52-69 heme oxygenase 1 Homo sapiens 94-98 35583593-0 2022 A Pretreatment with Isoorientin Attenuates Redox Disruption, Mitochondrial Impairment, and Inflammation Caused by Chlorpyrifos in a Dopaminergic Cell Line: Involvement of the Nrf2/HO-1 Axis. homoorientin 20-31 heme oxygenase 1 Homo sapiens 180-184 35583593-0 2022 A Pretreatment with Isoorientin Attenuates Redox Disruption, Mitochondrial Impairment, and Inflammation Caused by Chlorpyrifos in a Dopaminergic Cell Line: Involvement of the Nrf2/HO-1 Axis. Chlorpyrifos 114-126 heme oxygenase 1 Homo sapiens 180-184 35583593-8 2022 On the other hand, ISO upregulated HO-1 activity in SH-SY5Y cells. homoorientin 19-22 heme oxygenase 1 Homo sapiens 35-39 35583593-9 2022 Inhibition of HO-1 by zinc protoporphyrin-IX (ZnPP-IX) suppressed the cytoprotection induced by ISO in the CPF-treated cells. zinc protoporphyrin 22-44 heme oxygenase 1 Homo sapiens 14-18 35583593-9 2022 Inhibition of HO-1 by zinc protoporphyrin-IX (ZnPP-IX) suppressed the cytoprotection induced by ISO in the CPF-treated cells. zinc protoporphyrin 46-53 heme oxygenase 1 Homo sapiens 14-18 35583593-10 2022 Besides, silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) abolished the ISO-induced HO-1 upregulation and mitochondrial benefits induced by this flavone on the CPF-challenged cells. homoorientin 112-115 heme oxygenase 1 Homo sapiens 124-128 35583593-10 2022 Besides, silencing of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) abolished the ISO-induced HO-1 upregulation and mitochondrial benefits induced by this flavone on the CPF-challenged cells. flavone 185-192 heme oxygenase 1 Homo sapiens 124-128 35583593-11 2022 Thus, ISO protected mitochondria of the CPF-treated cells by an Nrf2/HO-1-dependent fashion in the SH-SY5Y cells. homoorientin 6-9 heme oxygenase 1 Homo sapiens 69-73 35620405-0 2022 11,12-Diacetyl-carnosol Protects SH-SY5Y Cells from Hydrogen Peroxide Damage through the Nrf2/HO-1 Pathway. 11,12-diacetyl-carnosol 0-23 heme oxygenase 1 Homo sapiens 94-98 35630697-2 2022 The newly designed and synthesized compounds are structurally related to nilotinib (NIL), a second-generation oral TKI, and to a series of imatinib (IM)-based TKIs, previously reported by our research group, these latter characterized by a hybrid structure between TKIs and heme oxygenase-1 (HO-1) inhibitors. nilotinib 73-82 heme oxygenase 1 Homo sapiens 274-290 35630697-2 2022 The newly designed and synthesized compounds are structurally related to nilotinib (NIL), a second-generation oral TKI, and to a series of imatinib (IM)-based TKIs, previously reported by our research group, these latter characterized by a hybrid structure between TKIs and heme oxygenase-1 (HO-1) inhibitors. nilotinib 73-82 heme oxygenase 1 Homo sapiens 292-296 35630697-2 2022 The newly designed and synthesized compounds are structurally related to nilotinib (NIL), a second-generation oral TKI, and to a series of imatinib (IM)-based TKIs, previously reported by our research group, these latter characterized by a hybrid structure between TKIs and heme oxygenase-1 (HO-1) inhibitors. Imatinib Mesylate 139-147 heme oxygenase 1 Homo sapiens 274-290 35630697-2 2022 The newly designed and synthesized compounds are structurally related to nilotinib (NIL), a second-generation oral TKI, and to a series of imatinib (IM)-based TKIs, previously reported by our research group, these latter characterized by a hybrid structure between TKIs and heme oxygenase-1 (HO-1) inhibitors. Imatinib Mesylate 139-147 heme oxygenase 1 Homo sapiens 292-296 35620405-9 2022 Conclusion: These results demonstrate that NO.20 protects SH-SY5Y cells from H2O2-induced neurotoxicity by activating the Nrf2/HO-1 pathway. Hydrogen Peroxide 77-81 heme oxygenase 1 Homo sapiens 127-131 35569637-0 2022 Synthesis and biological evaluation of scutellarein derivatives as neuroprotective agents via activating Nrf2/HO-1 pathway. scutellarein 39-51 heme oxygenase 1 Homo sapiens 110-114 35580917-9 2022 In addition, treatment with vinpocetine suppressed protein expression of Nrf2 and inhibited messenger RNA levels of heme oxygenase 1 and NAD(P)H dehydrogenase quinone 1 induced by cisplatin. vinpocetine 28-39 heme oxygenase 1 Homo sapiens 116-132 35580917-9 2022 In addition, treatment with vinpocetine suppressed protein expression of Nrf2 and inhibited messenger RNA levels of heme oxygenase 1 and NAD(P)H dehydrogenase quinone 1 induced by cisplatin. Cisplatin 180-189 heme oxygenase 1 Homo sapiens 116-132 35292342-0 2022 Preliminary studies of an imidazole-based alcohol derivative for imaging of Heme oxygenase 1. imidazole 26-35 heme oxygenase 1 Homo sapiens 76-92 35292342-0 2022 Preliminary studies of an imidazole-based alcohol derivative for imaging of Heme oxygenase 1. Alcohols 42-49 heme oxygenase 1 Homo sapiens 76-92 35292342-4 2022 We report herein the synthesis and characterization of a 11C-labeled imidazole-based alcohol derivative ((11C)QC-33) for imaging of HO-1 in the brain. Carbon-11 57-60 heme oxygenase 1 Homo sapiens 132-136 35292342-4 2022 We report herein the synthesis and characterization of a 11C-labeled imidazole-based alcohol derivative ((11C)QC-33) for imaging of HO-1 in the brain. imidazole 69-78 heme oxygenase 1 Homo sapiens 132-136 35292342-4 2022 We report herein the synthesis and characterization of a 11C-labeled imidazole-based alcohol derivative ((11C)QC-33) for imaging of HO-1 in the brain. Alcohols 85-92 heme oxygenase 1 Homo sapiens 132-136 35292342-4 2022 We report herein the synthesis and characterization of a 11C-labeled imidazole-based alcohol derivative ((11C)QC-33) for imaging of HO-1 in the brain. Carbon-11 106-109 heme oxygenase 1 Homo sapiens 132-136 35292342-7 2022 In vitro autoradiography studies indicated specific binding of (11C)QC-33 in the HO-1 rich regions, showing 75%, 75%, and 69% radioactivity binding reductions in cerebellum, brain stem, and midbrain, respectively. Carbon-11 64-67 heme oxygenase 1 Homo sapiens 81-85 35476430-3 2022 We found that the herbicidal-active imazethapyr enantiomer ((R)-IM) stimulated heme oxygenase-1 activity, triggered the release of the catalytic product Fe2+, increased reactive oxygen species production, decreased the DIMBOA content, and increased the DIMBOA-Fe content. imazethapyr enantiomer 36-58 heme oxygenase 1 Homo sapiens 79-95 35551540-1 2022 INTRODUCTION: Heme-oxygenase 1 (HMOX1) is a critical stress response gene that catalyzes the multistep oxidation of heme. Heme 116-120 heme oxygenase 1 Homo sapiens 14-30 35551540-1 2022 INTRODUCTION: Heme-oxygenase 1 (HMOX1) is a critical stress response gene that catalyzes the multistep oxidation of heme. Heme 116-120 heme oxygenase 1 Homo sapiens 32-37 35629355-7 2022 ROS interrupted the cellular oxidative balance by activating NOX and inhibiting HIF-1alpha and HO-1 expression. Reactive Oxygen Species 0-3 heme oxygenase 1 Homo sapiens 95-99 35601897-6 2022 The in vivo study showed that in situ injection of HT/HGA hydrogel significantly reduced malondialdehyde (MDA) production and increased glutathione (GSH) expression in lesion area after treatment for 3 or 21 days, which might be associated with the activation of Nrf2/HO-1 pathway. Glutathione 136-147 heme oxygenase 1 Homo sapiens 268-272 35592532-0 2022 Ferulic Acid Protects Human Lens Epithelial Cells against Ionizing Radiation-Induced Oxidative Damage by Activating Nrf2/HO-1 Signal Pathway. ferulic acid 0-12 heme oxygenase 1 Homo sapiens 121-125 35629355-8 2022 Pretreatment with N-acetylcysteine (NAC) reduced Apl-1-induced mitochondria-dependent apoptosis and preserved the expression of NOX, HO-1, and HIF-1a. Acetylcysteine 18-34 heme oxygenase 1 Homo sapiens 133-137 35629355-8 2022 Pretreatment with N-acetylcysteine (NAC) reduced Apl-1-induced mitochondria-dependent apoptosis and preserved the expression of NOX, HO-1, and HIF-1a. Acetylcysteine 36-39 heme oxygenase 1 Homo sapiens 133-137 35395341-7 2022 We established that dithianon (0.01-1 mumol.L-1) and captan (0.1, 1 mumol.L-1) induced mRNA expression of NQO1 and HMOX1 antioxidant enzymes. dithianone 20-29 heme oxygenase 1 Homo sapiens 115-120 35427872-10 2022 In contrast, honey"s anti-inflammatory actions and flavonoids induce anti-inflammatory and antioxidant pathways by inducing NRF2 target genes, including HO-1 and PRDX1. Flavonoids 51-61 heme oxygenase 1 Homo sapiens 153-157 35565605-8 2022 Additionally, dietary EA improved the antioxidant enzyme activity and mRNA levels of Nrf2/HO-1 in the ileum of heat-stressed broilers. Ellagic Acid 22-24 heme oxygenase 1 Homo sapiens 90-94 35305818-8 2022 As a potential mechanism, the expression of intracellular reactive oxygen species (ROS) and its related proteins, including p-ERK, NRF2, p-p38, HO-1, and gammaH2AX, was affected in EOC cells. Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 144-148 35015114-3 2022 RESULTS: In experimental models, curcumin showed its pleiotropic effects in retinal diseases like diabetic retinopathy by increasing anti-oxidant enzymes, upregulating HO-1, nrf2 and reducing or inhibiting inflammatory mediators, growth factors and by inhibiting proliferation and migration of retinal endothelial cells in a dose-dependent manner in HRPC, HREC and ARPE-19 cells. Curcumin 33-41 heme oxygenase 1 Homo sapiens 168-172 35015114-4 2022 In age-related macular degeneration, curcumin acts by reducing ROS and inhibiting apoptosis inducing proteins and cellular inflammatory genes and upregulating HO-1, thioredoxin and NQO1. Curcumin 37-45 heme oxygenase 1 Homo sapiens 159-163 35305818-8 2022 As a potential mechanism, the expression of intracellular reactive oxygen species (ROS) and its related proteins, including p-ERK, NRF2, p-p38, HO-1, and gammaH2AX, was affected in EOC cells. Reactive Oxygen Species 83-86 heme oxygenase 1 Homo sapiens 144-148 35428017-2 2022 The aim of this study was to investigate the role of heme in HO-1 and HO-2 induced colorectal carcinogenesis and the clinicopathological significance of their expressions in patients with colorectal carcinoma (CRC). Heme 53-57 heme oxygenase 1 Homo sapiens 61-65 35399331-0 2022 Carboxymethylated pachyman induces ferroptosis in ovarian cancer by suppressing NRF1/HO-1 signaling. pachyman 18-26 heme oxygenase 1 Homo sapiens 85-89 35428017-7 2022 HO-1 expression in SW480 was increased with all hemin concentrations and HO-2 expression was downregulated at the highest hemin concentration. Hemin 122-127 heme oxygenase 1 Homo sapiens 0-4 35428017-3 2022 METHODS: HO-1 and HO-2 expression alterations in normal colonic epithelial (FHC) and colon cancer cells (SW480) were explored following treatment with 0 microM, 25 microM, 100 microM and 250 microM concentrations of hemin, using qPCR. Hemin 216-221 heme oxygenase 1 Homo sapiens 9-13 35428017-12 2022 CONCLUSION: HO-1 and HO-2 expression is respectively induced and repressed by exogenous hemin in normal colon and colon cancer cells. Hemin 88-93 heme oxygenase 1 Homo sapiens 12-16 35428017-6 2022 RESULTS: Low concentrations of hemin caused upregulation and high concentration caused downregulation of HO-1 expression, whereas HO-2 expression was significantly downregulated with all hemin concentrations in FHC. Hemin 31-36 heme oxygenase 1 Homo sapiens 105-109 35428017-7 2022 HO-1 expression in SW480 was increased with all hemin concentrations and HO-2 expression was downregulated at the highest hemin concentration. Hemin 48-53 heme oxygenase 1 Homo sapiens 0-4 35306372-3 2022 Photochemical oxidation of isoprene leads to the formation of hydroperoxides, environmental oxidants that lead to inflammatory (IL-8) and adaptive (HMOX1) gene expression in human airway epithelial cells (HAEC). isoprene 27-35 heme oxygenase 1 Homo sapiens 148-153 35306372-3 2022 Photochemical oxidation of isoprene leads to the formation of hydroperoxides, environmental oxidants that lead to inflammatory (IL-8) and adaptive (HMOX1) gene expression in human airway epithelial cells (HAEC). Hydrogen Peroxide 62-76 heme oxygenase 1 Homo sapiens 148-153 35565370-3 2022 We quantified the heme-degrading enzyme, heme oxygenase-1 (HO-1, encoded by Hmox1) in normal peritoneum, endometriotic lesions and endometriosis-associated ovarian cancer (EAOC) of clear cell type (OCCC). Heme 18-22 heme oxygenase 1 Homo sapiens 41-57 35565370-3 2022 We quantified the heme-degrading enzyme, heme oxygenase-1 (HO-1, encoded by Hmox1) in normal peritoneum, endometriotic lesions and endometriosis-associated ovarian cancer (EAOC) of clear cell type (OCCC). Heme 18-22 heme oxygenase 1 Homo sapiens 59-63 35565370-3 2022 We quantified the heme-degrading enzyme, heme oxygenase-1 (HO-1, encoded by Hmox1) in normal peritoneum, endometriotic lesions and endometriosis-associated ovarian cancer (EAOC) of clear cell type (OCCC). Heme 18-22 heme oxygenase 1 Homo sapiens 76-81 35572500-10 2022 Further analysis showed that the Nrf2 inhibitor ML385 weakened TIPE2-induced activation of the Nrf2/HO-1 pathway, as well as TIPE2-induced suppression in M1 polarization of macrophage and inflammatory cytokines secretion. ML385 48-53 heme oxygenase 1 Homo sapiens 100-104 35490795-0 2022 Induction of ferroptosis by carnosic acid-mediated inactivation of Nrf2/HO-1 potentiates cisplatin responsiveness in OSCC cells. salvin 28-41 heme oxygenase 1 Homo sapiens 72-76 35490795-0 2022 Induction of ferroptosis by carnosic acid-mediated inactivation of Nrf2/HO-1 potentiates cisplatin responsiveness in OSCC cells. Cisplatin 89-98 heme oxygenase 1 Homo sapiens 72-76 35490795-9 2022 Furthermore, compared with parental cells, stronger activation of the Nrf2/HO-1/xCT signaling was observed in cisplatin-resistant cells, which was inhibited by carnosic acid. Cisplatin 110-119 heme oxygenase 1 Homo sapiens 75-79 35490795-9 2022 Furthermore, compared with parental cells, stronger activation of the Nrf2/HO-1/xCT signaling was observed in cisplatin-resistant cells, which was inhibited by carnosic acid. salvin 160-173 heme oxygenase 1 Homo sapiens 75-79 35490795-11 2022 Together, the current findings highlight that carnosic acid may re-sensitize cisplatin-resistant cells to cisplatin by inducing ferroptosis, which involves the inactivation of Nrf2/HO-1/xCT pathway. salvin 46-59 heme oxygenase 1 Homo sapiens 181-185 35490795-11 2022 Together, the current findings highlight that carnosic acid may re-sensitize cisplatin-resistant cells to cisplatin by inducing ferroptosis, which involves the inactivation of Nrf2/HO-1/xCT pathway. Cisplatin 77-86 heme oxygenase 1 Homo sapiens 181-185 35490795-11 2022 Together, the current findings highlight that carnosic acid may re-sensitize cisplatin-resistant cells to cisplatin by inducing ferroptosis, which involves the inactivation of Nrf2/HO-1/xCT pathway. Cisplatin 106-115 heme oxygenase 1 Homo sapiens 181-185 35426252-8 2022 Mechanically, EGCG upregulated nuclear factor erythroid 2-related factor 2 (Nrf2), oxygenase-1 (HO-1), and NADPH quinone oxidoreductase1 (NQO1). epigallocatechin gallate 14-18 heme oxygenase 1 Homo sapiens 96-100 35420033-8 2022 Sesamin significantly increased TNF-alpha/IL-4-induced superoxide dismutase (SOD), catalase (CAT), heme oxygenase 1 (HO-1), and nuclear factor erythroid 2 related factor 2 (Nrf2), and decreased malondialdehyde. sesamin 0-7 heme oxygenase 1 Homo sapiens 99-115 35420033-8 2022 Sesamin significantly increased TNF-alpha/IL-4-induced superoxide dismutase (SOD), catalase (CAT), heme oxygenase 1 (HO-1), and nuclear factor erythroid 2 related factor 2 (Nrf2), and decreased malondialdehyde. sesamin 0-7 heme oxygenase 1 Homo sapiens 117-121 35624663-7 2022 Furthermore, empagliflozin significantly upregulated the expressions of antioxidant enzymes superoxide dismutase (SOD) and HO-1. empagliflozin 13-26 heme oxygenase 1 Homo sapiens 123-127 35631320-0 2022 Activation of Nrf2/HO-1 Antioxidant Pathway by Heme Attenuates Calcification of Human Lens Epithelial Cells. Heme 47-51 heme oxygenase 1 Homo sapiens 19-23 35631320-8 2022 We used heme to activate Nrf2, which strongly upregulated the expression of Nrf2 and heme oxygenase-1 (HO-1). Heme 8-12 heme oxygenase 1 Homo sapiens 85-101 35631320-8 2022 We used heme to activate Nrf2, which strongly upregulated the expression of Nrf2 and heme oxygenase-1 (HO-1). Heme 8-12 heme oxygenase 1 Homo sapiens 103-107 35631320-11 2022 Anti-calcification effect of heme was lost when the transcriptional activity of Nrf2 or the enzyme activity of HO-1 was blocked with pharmacological inhibitors. Heme 29-33 heme oxygenase 1 Homo sapiens 111-115 35631320-12 2022 Among products of HO-1 catalyzed heme degradation iron mimicked the anti-calcification effect of heme. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 35631320-12 2022 Among products of HO-1 catalyzed heme degradation iron mimicked the anti-calcification effect of heme. Iron 50-54 heme oxygenase 1 Homo sapiens 18-22 35631320-12 2022 Among products of HO-1 catalyzed heme degradation iron mimicked the anti-calcification effect of heme. Heme 97-101 heme oxygenase 1 Homo sapiens 18-22 35631320-13 2022 We concluded that heme-induced upregulation of the Nrf2/HO-1 system inhibits HuLECs calcification through the liberation of heme iron. Heme 18-22 heme oxygenase 1 Homo sapiens 56-60 35631320-13 2022 We concluded that heme-induced upregulation of the Nrf2/HO-1 system inhibits HuLECs calcification through the liberation of heme iron. Heme 124-128 heme oxygenase 1 Homo sapiens 56-60 35631320-13 2022 We concluded that heme-induced upregulation of the Nrf2/HO-1 system inhibits HuLECs calcification through the liberation of heme iron. Iron 129-133 heme oxygenase 1 Homo sapiens 56-60 35422193-9 2022 Exo improved MI induced by SAP through activating Akt/Nrf2/HO-1 signaling pathway. BENSULIDE 27-30 heme oxygenase 1 Homo sapiens 59-63 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Heme 71-75 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Heme 71-75 heme oxygenase 1 Homo sapiens 18-22 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Carbon Monoxide 108-123 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Carbon Monoxide 108-123 heme oxygenase 1 Homo sapiens 18-22 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Carbon Monoxide 125-127 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Carbon Monoxide 125-127 heme oxygenase 1 Homo sapiens 18-22 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Biliverdine 130-140 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Biliverdine 130-140 heme oxygenase 1 Homo sapiens 18-22 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Biliverdine 142-144 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Biliverdine 142-144 heme oxygenase 1 Homo sapiens 18-22 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Iron 151-155 heme oxygenase 1 Homo sapiens 0-16 35624725-1 2022 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes the degradation of heme, releasing equimolar amounts of carbon monoxide (CO), biliverdin (BV), and iron. Iron 151-155 heme oxygenase 1 Homo sapiens 18-22 35624725-3 2022 However, iron constitutes an important product of HO-1 activity involved in the regulation of several cellular biological processes. Iron 9-13 heme oxygenase 1 Homo sapiens 50-54 35480872-0 2022 Biliverdin/Bilirubin Redox Pair Protects Lens Epithelial Cells against Oxidative Stress in Age-Related Cataract by Regulating NF-kappaB/iNOS and Nrf2/HO-1 Pathways. Biliverdine 0-10 heme oxygenase 1 Homo sapiens 150-154 35457199-6 2022 First, we observed an increase of HO-1 expression in Caco-2 cells treated with K-8-K and S-10-S, following the activation of the Keap1/Nrf2 pathway. k-8-k 79-84 heme oxygenase 1 Homo sapiens 34-38 35457199-6 2022 First, we observed an increase of HO-1 expression in Caco-2 cells treated with K-8-K and S-10-S, following the activation of the Keap1/Nrf2 pathway. S 10 89-95 heme oxygenase 1 Homo sapiens 34-38 35453467-0 2022 HO-1 Upregulation by Kaempferol via ROS-Dependent Nrf2-ARE Cascade Attenuates Lipopolysaccharide-Mediated Intercellular Cell Adhesion Molecule-1 Expression in Human Pulmonary Alveolar Epithelial Cells. kaempferol 21-31 heme oxygenase 1 Homo sapiens 0-4 35453467-0 2022 HO-1 Upregulation by Kaempferol via ROS-Dependent Nrf2-ARE Cascade Attenuates Lipopolysaccharide-Mediated Intercellular Cell Adhesion Molecule-1 Expression in Human Pulmonary Alveolar Epithelial Cells. Reactive Oxygen Species 36-39 heme oxygenase 1 Homo sapiens 0-4 35453467-2 2022 Heme oxygenase-1 (HO-1) is a key antioxidant enzyme that could be induced by kaempferol (KPR) and exerts anti-inflammatory effects. kaempferol 77-87 heme oxygenase 1 Homo sapiens 0-16 35453467-2 2022 Heme oxygenase-1 (HO-1) is a key antioxidant enzyme that could be induced by kaempferol (KPR) and exerts anti-inflammatory effects. kaempferol 77-87 heme oxygenase 1 Homo sapiens 18-22 35453467-2 2022 Heme oxygenase-1 (HO-1) is a key antioxidant enzyme that could be induced by kaempferol (KPR) and exerts anti-inflammatory effects. kaempferol 89-92 heme oxygenase 1 Homo sapiens 0-16 35453467-2 2022 Heme oxygenase-1 (HO-1) is a key antioxidant enzyme that could be induced by kaempferol (KPR) and exerts anti-inflammatory effects. kaempferol 89-92 heme oxygenase 1 Homo sapiens 18-22 35496911-0 2022 Heme Oxygenase-1 Protects Hair Cells From Gentamicin-Induced Death. Gentamicins 42-52 heme oxygenase 1 Homo sapiens 0-16 35496911-4 2022 We found that during gentamicin-induced death in HCs, Heme oxygenase-1 (HO-1) had a high fold change in the HCs of the cochlea. Gentamicins 21-31 heme oxygenase 1 Homo sapiens 54-70 35496911-4 2022 We found that during gentamicin-induced death in HCs, Heme oxygenase-1 (HO-1) had a high fold change in the HCs of the cochlea. Gentamicins 21-31 heme oxygenase 1 Homo sapiens 72-76 35496911-5 2022 Moreover, the use of CoPPIX to induce HO-1 inhibited gentamicin-induced HC death, while HO-1 inhibitors ZnPPIX after CoPPIX reversed this process. Gentamicins 53-63 heme oxygenase 1 Homo sapiens 38-42 35496911-5 2022 Moreover, the use of CoPPIX to induce HO-1 inhibited gentamicin-induced HC death, while HO-1 inhibitors ZnPPIX after CoPPIX reversed this process. zinc protoporphyrin 104-110 heme oxygenase 1 Homo sapiens 38-42 35496911-5 2022 Moreover, the use of CoPPIX to induce HO-1 inhibited gentamicin-induced HC death, while HO-1 inhibitors ZnPPIX after CoPPIX reversed this process. zinc protoporphyrin 104-110 heme oxygenase 1 Homo sapiens 88-92 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 142-152 heme oxygenase 1 Homo sapiens 87-91 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 142-152 heme oxygenase 1 Homo sapiens 131-135 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 142-152 heme oxygenase 1 Homo sapiens 184-188 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 209-219 heme oxygenase 1 Homo sapiens 87-91 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 209-219 heme oxygenase 1 Homo sapiens 131-135 35496911-6 2022 Furthermore, the inhibitors of NF-E2-related factor-2 (Nrf2) reduced the expression of HO-1 and inhibited the protective effect of HO-1 after gentamicin, thus suggesting that the Nrf2/HO-1 axis might regulate gentamicin-associated ototoxicity. Gentamicins 209-219 heme oxygenase 1 Homo sapiens 184-188 35496911-8 2022 In summary, these findings indicated that HO-1 protects HCs from gentamicin by up-regulating its expression in HCs and interacting with Nrf2 to inhibit reactive oxygen species (ROS). Gentamicins 65-75 heme oxygenase 1 Homo sapiens 42-46 35496911-8 2022 In summary, these findings indicated that HO-1 protects HCs from gentamicin by up-regulating its expression in HCs and interacting with Nrf2 to inhibit reactive oxygen species (ROS). Reactive Oxygen Species 152-175 heme oxygenase 1 Homo sapiens 42-46 35496911-8 2022 In summary, these findings indicated that HO-1 protects HCs from gentamicin by up-regulating its expression in HCs and interacting with Nrf2 to inhibit reactive oxygen species (ROS). Reactive Oxygen Species 177-180 heme oxygenase 1 Homo sapiens 42-46 35453452-0 2022 TLR4 Signaling and Heme Oxygenase-1/Carbon Monoxide Pathway Crosstalk Induces Resiliency of Myeloma Plasma Cells to Bortezomib Treatment. Bortezomib 116-126 heme oxygenase 1 Homo sapiens 19-35 35453452-2 2022 Since our group previously demonstrated that heme oxygenase-1 (HO-1) and Toll-like receptor 4 (TLR4) are two signaling pathways protecting MM cells from the proteasome inhibitor bortezomib (BTZ), we here evaluated their cross-regulation by a pharmacological approach. Bortezomib 178-188 heme oxygenase 1 Homo sapiens 45-61 35453452-2 2022 Since our group previously demonstrated that heme oxygenase-1 (HO-1) and Toll-like receptor 4 (TLR4) are two signaling pathways protecting MM cells from the proteasome inhibitor bortezomib (BTZ), we here evaluated their cross-regulation by a pharmacological approach. Bortezomib 178-188 heme oxygenase 1 Homo sapiens 63-67 35453452-2 2022 Since our group previously demonstrated that heme oxygenase-1 (HO-1) and Toll-like receptor 4 (TLR4) are two signaling pathways protecting MM cells from the proteasome inhibitor bortezomib (BTZ), we here evaluated their cross-regulation by a pharmacological approach. Bortezomib 190-193 heme oxygenase 1 Homo sapiens 45-61 35453452-2 2022 Since our group previously demonstrated that heme oxygenase-1 (HO-1) and Toll-like receptor 4 (TLR4) are two signaling pathways protecting MM cells from the proteasome inhibitor bortezomib (BTZ), we here evaluated their cross-regulation by a pharmacological approach. Bortezomib 190-193 heme oxygenase 1 Homo sapiens 63-67 35453452-3 2022 We found that cell toxicity and mitochondrial depolarization by BTZ were increased upon inhibition of HO-1 and TLR4 by using tin protoporphyrin IX (SnPP) and TAK-242, respectively. Bortezomib 64-67 heme oxygenase 1 Homo sapiens 102-106 35453452-3 2022 We found that cell toxicity and mitochondrial depolarization by BTZ were increased upon inhibition of HO-1 and TLR4 by using tin protoporphyrin IX (SnPP) and TAK-242, respectively. tin protoporphyrin IX 125-146 heme oxygenase 1 Homo sapiens 102-106 35453452-3 2022 We found that cell toxicity and mitochondrial depolarization by BTZ were increased upon inhibition of HO-1 and TLR4 by using tin protoporphyrin IX (SnPP) and TAK-242, respectively. S-Nitroso-N-propionyl-D,L-penicillamine 148-152 heme oxygenase 1 Homo sapiens 102-106 35453452-3 2022 We found that cell toxicity and mitochondrial depolarization by BTZ were increased upon inhibition of HO-1 and TLR4 by using tin protoporphyrin IX (SnPP) and TAK-242, respectively. ethyl 6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex-1-ene-1-carboxylate 158-165 heme oxygenase 1 Homo sapiens 102-106 35453452-4 2022 Furthermore, the combination of TAK-242 and BTZ activated mitophagy and decreased the unfolded protein response (UPR) survival pathway in association with a downregulation in HO-1 expression. ethyl 6-(N-(2-chloro-4-fluorophenyl)sulfamoyl)cyclohex-1-ene-1-carboxylate 32-39 heme oxygenase 1 Homo sapiens 175-179 35453452-4 2022 Furthermore, the combination of TAK-242 and BTZ activated mitophagy and decreased the unfolded protein response (UPR) survival pathway in association with a downregulation in HO-1 expression. Bortezomib 44-47 heme oxygenase 1 Homo sapiens 175-179 35453452-6 2022 Interestingly, treatment of cells with either hemin, an HO-1 inducer, or supplementation with carbon monoxide (CO), a by-product of HO-1 enzymatic activity, increased TLR4 expression. Hemin 46-51 heme oxygenase 1 Homo sapiens 56-60 35453452-6 2022 Interestingly, treatment of cells with either hemin, an HO-1 inducer, or supplementation with carbon monoxide (CO), a by-product of HO-1 enzymatic activity, increased TLR4 expression. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 132-136 35453452-6 2022 Interestingly, treatment of cells with either hemin, an HO-1 inducer, or supplementation with carbon monoxide (CO), a by-product of HO-1 enzymatic activity, increased TLR4 expression. Carbon Monoxide 111-113 heme oxygenase 1 Homo sapiens 132-136 35453452-7 2022 In conclusion, we showed that treatment of MM cells with BTZ triggers the TLR4/HO-1/CO axis, serving as a stress-responsive signal that leads to increased cell survival while protecting mitochondria against BTZ and ultimately promoting drug resistance. Bortezomib 57-60 heme oxygenase 1 Homo sapiens 79-83 35480872-0 2022 Biliverdin/Bilirubin Redox Pair Protects Lens Epithelial Cells against Oxidative Stress in Age-Related Cataract by Regulating NF-kappaB/iNOS and Nrf2/HO-1 Pathways. Bilirubin 11-20 heme oxygenase 1 Homo sapiens 150-154 35453452-7 2022 In conclusion, we showed that treatment of MM cells with BTZ triggers the TLR4/HO-1/CO axis, serving as a stress-responsive signal that leads to increased cell survival while protecting mitochondria against BTZ and ultimately promoting drug resistance. Bortezomib 207-210 heme oxygenase 1 Homo sapiens 79-83 35480872-12 2022 These findings implicated that BV/BR redox pair protects LECs against H2O2-induced apoptosis by regulating NF-kappaB/iNOS and Nrf2/HO-1 pathways. Hydrogen Peroxide 70-74 heme oxygenase 1 Homo sapiens 131-135 35396773-6 2022 We screened for a novel chlorambucil-polyamide conjugate (Chb) to target PRCC-TFE3 dependent transcription, and identified Chb16 as a PRCC-TFE3-dependent transcriptional inhibitor of HMOX1 expression. Chlorambucil 24-36 heme oxygenase 1 Homo sapiens 183-188 35396773-6 2022 We screened for a novel chlorambucil-polyamide conjugate (Chb) to target PRCC-TFE3 dependent transcription, and identified Chb16 as a PRCC-TFE3-dependent transcriptional inhibitor of HMOX1 expression. Nylons 37-46 heme oxygenase 1 Homo sapiens 183-188 35393708-0 2022 Heme oxygenase-1 induction by gallic acid-g-chitosan is an important event in modulating adipocyte differentiation. Gallic Acid 30-41 heme oxygenase 1 Homo sapiens 0-16 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. Gallic Acid 76-87 heme oxygenase 1 Homo sapiens 152-168 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. Gallic Acid 76-87 heme oxygenase 1 Homo sapiens 170-174 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. g-chitosan 88-98 heme oxygenase 1 Homo sapiens 152-168 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. g-chitosan 88-98 heme oxygenase 1 Homo sapiens 170-174 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. gallic acid-g-chitosan 100-103 heme oxygenase 1 Homo sapiens 152-168 35393708-4 2022 This study focused on investigating the probable anti-adipogenic effects of gallic acid-g-chitosan (GAC) and plain chitosan (PC) through regulating the heme oxygenase-1 (HO-1)/Nrf2 pathway on mesenchymal stem cells. gallic acid-g-chitosan 100-103 heme oxygenase 1 Homo sapiens 170-174 35393708-7 2022 On the contrary, zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor reversed these effects of GAC on adipogenesis. zinc protoporphyrin 17-39 heme oxygenase 1 Homo sapiens 50-54 35393708-7 2022 On the contrary, zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor reversed these effects of GAC on adipogenesis. zinc protoporphyrin 41-45 heme oxygenase 1 Homo sapiens 50-54 35393708-8 2022 Taken all together, this study revealed that grafting GA onto the chitosan improved potential anti-adipogenic activity by induction of the HO-1/Nrf2 pathway on mesenchymal stem cells (MSCs). Gallium 54-56 heme oxygenase 1 Homo sapiens 139-143 35464033-4 2022 Additionally, EBCs and NEBCs from G. rubra displayed anti-inflammatory functions via inhibiting the production of nitric oxide (NO), reactive oxygen species (ROS), and proinflammatory cytokines in activated macrophages and regulating the expression levels of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), NADPH-quinone oxidoreductase-1 (NQO-1), and heme oxygenase 1 (HO-1). ebcs 14-18 heme oxygenase 1 Homo sapiens 369-385 35432723-10 2022 sip62 interference results showed that GDN cause the less expression of HO-1 and Keap1 and also fail to inhibit cytokines after sip62 interference. sip62 0-5 heme oxygenase 1 Homo sapiens 72-76 35464033-4 2022 Additionally, EBCs and NEBCs from G. rubra displayed anti-inflammatory functions via inhibiting the production of nitric oxide (NO), reactive oxygen species (ROS), and proinflammatory cytokines in activated macrophages and regulating the expression levels of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), NADPH-quinone oxidoreductase-1 (NQO-1), and heme oxygenase 1 (HO-1). ebcs 14-18 heme oxygenase 1 Homo sapiens 387-391 35453402-6 2022 Transient knockdown or inhibitor-based suppression of HO-1 enhances vulnerability to the TKI, quizartinib, in both TKI-resistant and sensitive primary AML and cell line models. quizartinib 94-105 heme oxygenase 1 Homo sapiens 54-58 35453402-7 2022 NRF2 suppression (genetically or pharmacologically using brusatol) results in decreased HO-1, suggesting that TKI-resistance is dependent on an active NRF2-driven pathway. brusatol 57-65 heme oxygenase 1 Homo sapiens 88-92 35422894-0 2022 Choline Protects the Heart from Doxorubicin-Induced Cardiotoxicity through Vagal Activation and Nrf2/HO-1 Pathway. Choline 0-7 heme oxygenase 1 Homo sapiens 101-105 35422894-0 2022 Choline Protects the Heart from Doxorubicin-Induced Cardiotoxicity through Vagal Activation and Nrf2/HO-1 Pathway. Doxorubicin 32-43 heme oxygenase 1 Homo sapiens 101-105 35422894-5 2022 Levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1), which are antioxidant markers, were lowered by DOX and upregulated by choline. Doxorubicin 137-140 heme oxygenase 1 Homo sapiens 65-81 35422894-5 2022 Levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1), which are antioxidant markers, were lowered by DOX and upregulated by choline. Doxorubicin 137-140 heme oxygenase 1 Homo sapiens 83-87 35422894-5 2022 Levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1), which are antioxidant markers, were lowered by DOX and upregulated by choline. Choline 160-167 heme oxygenase 1 Homo sapiens 65-81 35422894-5 2022 Levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme-oxygenase-1 (HO-1), which are antioxidant markers, were lowered by DOX and upregulated by choline. Choline 160-167 heme oxygenase 1 Homo sapiens 83-87 35218740-10 2022 Taken together, the present study indicates that CORM-2-induced Nrf2/HO-1 alleviates IL-6/Jak2/Stat3-mediated inflammatory responses to Ang II by inhibiting NADPH oxidase- and mitochondria-derived ROS, suggesting that CORM-2 is a promising pharmacologic candidate to reverse the pathological changes involved in the inflammation of vessel wall for the prevention and treatment of AAA. ros 197-200 heme oxygenase 1 Homo sapiens 69-73 35408731-6 2022 It could reduce UV-induced intracellular ROS generation and initiate the antioxidant defense system by activating the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway in human skin fibroblasts. Reactive Oxygen Species 41-44 heme oxygenase 1 Homo sapiens 169-185 35408731-6 2022 It could reduce UV-induced intracellular ROS generation and initiate the antioxidant defense system by activating the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway in human skin fibroblasts. Reactive Oxygen Species 41-44 heme oxygenase 1 Homo sapiens 187-191 35366876-11 2022 PRM verified that the apoptosis-related proteins HMOX1 and GCLM were up-regulated and IL1B was down-regulated in BCPAP cells treated with parthenolide. parthenolide 138-150 heme oxygenase 1 Homo sapiens 49-54 35379891-5 2022 The network analyses led to the construction of the respiratory system-related biological network associated with formaldehyde exposure, and six upregulated hub genes (AREG, CXCL2, HMOX1, PLAUR, PTGS2, and TIMP1) were identified. Formaldehyde 114-126 heme oxygenase 1 Homo sapiens 181-186 35092472-0 2022 Astragaloside IV suppresses migration and invasion of TGF-beta1-induced human hepatoma HuH-7 cells by regulating Nrf2/HO-1 and TGF-beta1/Smad3 pathways. astragaloside 0-13 heme oxygenase 1 Homo sapiens 118-122 35350102-10 2022 Furthermore, spermine enhanced Nrf2 nuclear translocation, thereby increasing heme oxygenase-1 and NADPH quinone oxidoreductase-1 expression, which subsequently reduced ROS production. Spermine 13-21 heme oxygenase 1 Homo sapiens 78-94 35361044-0 2022 Curcumin analog, GO-Y078, induces HO-1 transactivation-mediated apoptotic cell death of oral cancer cells by triggering MAPK pathways and AP-1 DNA-binding activity. Curcumin 0-8 heme oxygenase 1 Homo sapiens 34-38 35361044-0 2022 Curcumin analog, GO-Y078, induces HO-1 transactivation-mediated apoptotic cell death of oral cancer cells by triggering MAPK pathways and AP-1 DNA-binding activity. GO-Y078 17-24 heme oxygenase 1 Homo sapiens 34-38 35361044-5 2022 GO-Y078 transcriptionally induced upregulation of the HO-1 gene by increasing the AP-1 DNA-binding activity, which was initiated by activation of the p38 /JNK1/2 pathways. GO-Y078 0-7 heme oxygenase 1 Homo sapiens 54-58 35531734-12 2022 FLFs showed a protective effect against H_2O_2-induced oxidative damage in L02 cells, and the underlying mechanism is associated with the enhancement of cell capability in clearing oxygen free radicals and the inhibition of apoptosis by the activation of the MAPKs/Nrf2/HO-1 signaling pathway. h_2o 40-44 heme oxygenase 1 Homo sapiens 270-274 35396005-3 2022 Heme oxygenase 1 (HO 1) is a key rate limiting enzyme in the process of heme metabolism, which has the functions of anti inflammation, anti oxidation, anti apoptosis, anti aging, reducing cell damage and promoting angiogenesis. Heme 72-76 heme oxygenase 1 Homo sapiens 0-16 35396005-3 2022 Heme oxygenase 1 (HO 1) is a key rate limiting enzyme in the process of heme metabolism, which has the functions of anti inflammation, anti oxidation, anti apoptosis, anti aging, reducing cell damage and promoting angiogenesis. Heme 72-76 heme oxygenase 1 Homo sapiens 18-22 35192145-8 2022 TG caused the increases in intracellular reactive oxygen species (ROS) and in the protein levels of pSer40-Nrf2 and hemoglobin oxygenase 1 (HO-1). Thapsigargin 0-2 heme oxygenase 1 Homo sapiens 116-138 35192145-8 2022 TG caused the increases in intracellular reactive oxygen species (ROS) and in the protein levels of pSer40-Nrf2 and hemoglobin oxygenase 1 (HO-1). Thapsigargin 0-2 heme oxygenase 1 Homo sapiens 140-144 35085765-9 2022 Additionally, photo-activated DMA upregulated the oxidative stress marker gene, hemeoxygenase-1 and apoptotic marker genes (cytochrome-C, caspase-3, caspase-9). N-myristoyl-alaninol 30-33 heme oxygenase 1 Homo sapiens 80-95 35419025-8 2022 Finally, Hemin, the agonist of HO-1, was applied in rescue assays, thereby verifying the mechanism of KLF7 modulating osteoclast differentiation by HO-1. Hemin 9-14 heme oxygenase 1 Homo sapiens 31-35 35453348-4 2022 Increased levels of Reactive Oxygen Species (ROS) activate NRF2 signalling, inducing the expression of antioxidant enzymes, such as haem oxygenase (HO-1), catalase (CAT), glutathione peroxidase (GPx) and superoxide dismutase (SOD), that protect cells against oxidative stress. Reactive Oxygen Species 20-43 heme oxygenase 1 Homo sapiens 148-152 35453348-4 2022 Increased levels of Reactive Oxygen Species (ROS) activate NRF2 signalling, inducing the expression of antioxidant enzymes, such as haem oxygenase (HO-1), catalase (CAT), glutathione peroxidase (GPx) and superoxide dismutase (SOD), that protect cells against oxidative stress. Reactive Oxygen Species 45-48 heme oxygenase 1 Homo sapiens 148-152 35419301-2 2022 HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. Heme 15-19 heme oxygenase 1 Homo sapiens 0-4 35419301-2 2022 HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 0-4 35419301-2 2022 HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 87-89 heme oxygenase 1 Homo sapiens 0-4 35419301-2 2022 HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. Biliverdine 92-102 heme oxygenase 1 Homo sapiens 0-4 35419301-2 2022 HO-1 catalyzes heme degradation, which gives rise to the formation of carbon monoxide (CO), biliverdin, and iron. Iron 108-112 heme oxygenase 1 Homo sapiens 0-4 35419301-3 2022 The upregulation of HO-1 under pathological conditions associated with cellular stress represents an important cytoprotective defense mechanism by virtue of the anti-oxidant properties of the bilirubin and the anti-inflammatory effect of the CO produced. Bilirubin 192-201 heme oxygenase 1 Homo sapiens 20-24 35453313-0 2022 Reduced Levels of H2S in Diabetes-Associated Osteoarthritis Are Linked to Hyperglycaemia, Nrf-2/HO-1 Signalling Downregulation and Chondrocyte Dysfunction. Deuterium 18-21 heme oxygenase 1 Homo sapiens 96-100 35388300-9 2022 Codonopsis pilosula could upregulate HMOX1 via luteolin, capsaicin, and sulforaphane. Capsaicin 57-66 heme oxygenase 1 Homo sapiens 37-42 35388300-9 2022 Codonopsis pilosula could upregulate HMOX1 via luteolin, capsaicin, and sulforaphane. sulforaphane 72-84 heme oxygenase 1 Homo sapiens 37-42 35453313-6 2022 In addition, our results indicate that H2S is an inductor of the Nrf-2/HO-1 signalling pathway in cartilage, but is also a downstream target of Nrf-2 transcriptional activity. Deuterium 39-42 heme oxygenase 1 Homo sapiens 71-75 35259284-8 2022 Furthermore, the NCHDH and NTHDH treatment significantly enhanced the antioxidants signaling proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1). nchdh 17-22 heme oxygenase 1 Homo sapiens 165-181 35332611-4 2022 The expression level of nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 markedly increased after BE treatment will intimidate A549 cells proliferation by the ROS-independent pathway via the antioxidant pathway. baicalein 114-116 heme oxygenase 1 Homo sapiens 72-88 35332611-4 2022 The expression level of nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 markedly increased after BE treatment will intimidate A549 cells proliferation by the ROS-independent pathway via the antioxidant pathway. Reactive Oxygen Species 175-178 heme oxygenase 1 Homo sapiens 72-88 35401165-5 2022 The in vitro results showed that: 1) Fr increased the expression of antioxidant enzymes including SOD1 and HO-1 to inhibit high glucose (HG)-induced fibronectin (FN) and inflammatory cell adhesion molecule (ICAM-1) overexpression; 2) Fr exerted antioxidant effect through activating the Nrf2/ARE pathway; 3) Fr significantly up-regulated the expression of Cx43 in HG-induced glomerular mesangial cells (GMCs), while the knock down of Cx43 largely impaired the activation of Nrf2/ARE pathway induced by Fr; 4) Fr promoted the activation of Nrf2/ARE pathway via regulating the interaction between Cx43 and AKT. Glucose 128-135 heme oxygenase 1 Homo sapiens 107-111 35387349-0 2022 Metformin Promotes Differentiation and Attenuates H2O2-Induced Oxidative Damage of Osteoblasts via the PI3K/AKT/Nrf2/HO-1 Pathway. Metformin 0-9 heme oxygenase 1 Homo sapiens 117-121 35387349-0 2022 Metformin Promotes Differentiation and Attenuates H2O2-Induced Oxidative Damage of Osteoblasts via the PI3K/AKT/Nrf2/HO-1 Pathway. Hydrogen Peroxide 50-54 heme oxygenase 1 Homo sapiens 117-121 35387349-9 2022 Antioxidative Nrf2/HO-1 pathway, regarded as the downstream of PI3K/AKT pathway, was modulated by metformin in the protective process. Metformin 98-107 heme oxygenase 1 Homo sapiens 19-23 35387349-11 2022 In conclusion, our study revealed that metformin promoted osteogenic differentiation and H2O2-induced oxidative damage of osteoblasts via the PI3K/AKT/Nrf2/HO-1 pathway. Metformin 39-48 heme oxygenase 1 Homo sapiens 156-160 35387349-11 2022 In conclusion, our study revealed that metformin promoted osteogenic differentiation and H2O2-induced oxidative damage of osteoblasts via the PI3K/AKT/Nrf2/HO-1 pathway. Hydrogen Peroxide 89-93 heme oxygenase 1 Homo sapiens 156-160 35286219-6 2022 Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of ROS through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-Nrf2-HO-1 axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. Raloxifene Hydrochloride 17-27 heme oxygenase 1 Homo sapiens 201-205 35286219-6 2022 Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of ROS through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-Nrf2-HO-1 axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. ros 105-108 heme oxygenase 1 Homo sapiens 201-205 35286219-6 2022 Mechanistically, raloxifene suppressed NLRP3 inflammasomes activation by lowering the cellular levels of ROS through the modulation of redox signaling mediated via aryl hydrocarbon receptor (AhR)-Nrf2-HO-1 axis or the impaired generation of mitochondrial ROS in a mitophagy-dependent manner. ros 255-258 heme oxygenase 1 Homo sapiens 201-205 35453311-6 2022 These results indicated that luteolin treatment retarded oxaliplatin-induced tumor growth by facilitating apoptotic cell death and inhibiting HO-1-mediated cytoprotection. Oxaliplatin 57-68 heme oxygenase 1 Homo sapiens 142-146 35300705-13 2022 Moreover, the encapsulation of glial cell-derived neurotrophic factor (GNDF)-containing DHAH-NLCs (DHAH-NLC-GNDF) activated the Nrf2/HO-1 pathway, suggesting the triggering of the endogenous anti-oxidative system present in microglia. dhah 88-92 heme oxygenase 1 Homo sapiens 133-137 35300705-13 2022 Moreover, the encapsulation of glial cell-derived neurotrophic factor (GNDF)-containing DHAH-NLCs (DHAH-NLC-GNDF) activated the Nrf2/HO-1 pathway, suggesting the triggering of the endogenous anti-oxidative system present in microglia. dhah 99-103 heme oxygenase 1 Homo sapiens 133-137 35260560-7 2022 Further findings revealed that HO-1 was essential for osteocyte ferroptosis in DOP and that its promoter activity was controlled by the interaction between the upstream NRF2 and c-JUN transcription factors. Diethylhexyl Phthalate 79-82 heme oxygenase 1 Homo sapiens 31-35 35260560-8 2022 Targeting ferroptosis or HO-1 efficiently rescued osteocyte death in DOP by disrupting the vicious cycle between lipid peroxidation and HO-1 activation, eventually ameliorating trabecular deterioration. Diethylhexyl Phthalate 69-72 heme oxygenase 1 Homo sapiens 25-29 35260560-8 2022 Targeting ferroptosis or HO-1 efficiently rescued osteocyte death in DOP by disrupting the vicious cycle between lipid peroxidation and HO-1 activation, eventually ameliorating trabecular deterioration. Diethylhexyl Phthalate 69-72 heme oxygenase 1 Homo sapiens 136-140 35350242-9 2022 Furthermore, knockdown of GSK3beta significantly reduced nobiletin-induced ferroptosis and upregulated the Keap1/Nrf2/HO-1 signalling pathway, while the opposite was observed in cells overexpressing GSK3beta. nobiletin 57-66 heme oxygenase 1 Homo sapiens 118-122 35259284-8 2022 Furthermore, the NCHDH and NTHDH treatment significantly enhanced the antioxidants signaling proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1). nchdh 17-22 heme oxygenase 1 Homo sapiens 183-187 35300173-0 2022 Local Treatment of Hydrogen-Rich Saline Promotes Wound Healing In Vivo by Inhibiting Oxidative Stress via Nrf-2/HO-1 Pathway. Hydrogen 19-27 heme oxygenase 1 Homo sapiens 112-116 35259284-8 2022 Furthermore, the NCHDH and NTHDH treatment significantly enhanced the antioxidants signaling proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1). nthdh 27-32 heme oxygenase 1 Homo sapiens 165-181 35300173-0 2022 Local Treatment of Hydrogen-Rich Saline Promotes Wound Healing In Vivo by Inhibiting Oxidative Stress via Nrf-2/HO-1 Pathway. Sodium Chloride 33-39 heme oxygenase 1 Homo sapiens 112-116 35300173-11 2022 Besides, hydrogen-rich medium relieved the oxidative stress via the activation of the Nrf-2/heme oxygenase-1 (HO-1) pathway. Hydrogen 9-17 heme oxygenase 1 Homo sapiens 92-108 35300173-11 2022 Besides, hydrogen-rich medium relieved the oxidative stress via the activation of the Nrf-2/heme oxygenase-1 (HO-1) pathway. Hydrogen 9-17 heme oxygenase 1 Homo sapiens 110-114 35259284-8 2022 Furthermore, the NCHDH and NTHDH treatment significantly enhanced the antioxidants signaling proteins such as nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase 1 (HO-1). nthdh 27-32 heme oxygenase 1 Homo sapiens 183-187 35300173-13 2022 Hydrogen relieves the oxidative stress in the wound microenvironment via Nrf-2/HO-1 signaling pathway. Hydrogen 0-8 heme oxygenase 1 Homo sapiens 79-83 35281466-1 2022 Recently, we found that 5,8-dihydroxy-4",7-dimethoxyflavone (DDF) upregulated the expression of heme oxygenase (HO)-1 via p38 mitogen-activated protein kinase/nuclear factor-erythroid factor 2-related factor 2 (MAPK/Nrf2) pathway in human cardiac fibroblasts (HCFs). 5,8-dihydroxy-4",7-dimethoxyflavone 24-59 heme oxygenase 1 Homo sapiens 96-117 35179529-5 2022 Under hypoxia, the expression of Nrf2 and heme oxygenase-1 was up-regulated by SFN treatment. sulforaphane 79-82 heme oxygenase 1 Homo sapiens 42-58 35281466-1 2022 Recently, we found that 5,8-dihydroxy-4",7-dimethoxyflavone (DDF) upregulated the expression of heme oxygenase (HO)-1 via p38 mitogen-activated protein kinase/nuclear factor-erythroid factor 2-related factor 2 (MAPK/Nrf2) pathway in human cardiac fibroblasts (HCFs). ddf 61-64 heme oxygenase 1 Homo sapiens 96-117 35281466-11 2022 Chromatin immunoprecipitation assays revealed the interaction between Sp1 and the binding site of proximal ARE on the HO-1 promoter, which was abolished by glutathione, AG1478, Go6976, LY294002, or mithramycin A. Glutathione 156-167 heme oxygenase 1 Homo sapiens 118-122 35281466-11 2022 Chromatin immunoprecipitation assays revealed the interaction between Sp1 and the binding site of proximal ARE on the HO-1 promoter, which was abolished by glutathione, AG1478, Go6976, LY294002, or mithramycin A. RTKI cpd 169-175 heme oxygenase 1 Homo sapiens 118-122 35281466-11 2022 Chromatin immunoprecipitation assays revealed the interaction between Sp1 and the binding site of proximal ARE on the HO-1 promoter, which was abolished by glutathione, AG1478, Go6976, LY294002, or mithramycin A. Go 6976 177-183 heme oxygenase 1 Homo sapiens 118-122 35281466-11 2022 Chromatin immunoprecipitation assays revealed the interaction between Sp1 and the binding site of proximal ARE on the HO-1 promoter, which was abolished by glutathione, AG1478, Go6976, LY294002, or mithramycin A. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 185-193 heme oxygenase 1 Homo sapiens 118-122 35281466-11 2022 Chromatin immunoprecipitation assays revealed the interaction between Sp1 and the binding site of proximal ARE on the HO-1 promoter, which was abolished by glutathione, AG1478, Go6976, LY294002, or mithramycin A. mithramycin A 198-211 heme oxygenase 1 Homo sapiens 118-122 35281466-13 2022 Pretreatment with an inhibitor of HO-1: zinc protoporphyrin IX reversed these inhibitory effects of HO-1. zinc protoporphyrin 40-62 heme oxygenase 1 Homo sapiens 34-38 35309045-8 2022 We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Glutathione 18-29 heme oxygenase 1 Homo sapiens 100-104 35309045-8 2022 We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Glutathione 39-50 heme oxygenase 1 Homo sapiens 100-104 35309045-8 2022 We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Glutathione 52-55 heme oxygenase 1 Homo sapiens 100-104 35309045-8 2022 We concluded that glutathione/oxidized glutathione (GSH/GSSG) conversion involved the PI3K/Akt-Nrf2/HO-1 signaling pathway and that the antioxidant enzyme-mediated mitochondrial apoptosis pathway of osteoblasts was necessary for the development of postmenopausal osteoporosis. Glutathione Disulfide 56-60 heme oxygenase 1 Homo sapiens 100-104 35281466-13 2022 Pretreatment with an inhibitor of HO-1: zinc protoporphyrin IX reversed these inhibitory effects of HO-1. zinc protoporphyrin 40-62 heme oxygenase 1 Homo sapiens 100-104 35024467-5 2022 Meanwhile, PEITC treatment ameliorated DON-induced an increase of the inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) mRNA levels and intracellular reactive oxygen species (ROS) level, and a decrease of glutathione peroxidase 1 (GPx1), superoxide dismutase 2 (SOD2), catalase (CAT) and heme oxygenase 1 (HO-1) mRNA levels. phenethyl isothiocyanate 11-16 heme oxygenase 1 Homo sapiens 306-322 35510228-7 2022 High dose of quercetin significantly decreased the gene expressions of NF-kappaB and TNFalpha, whereas the alternations of Nrf2 and HO-1 gene expressions were not significant in quercetin groups in compared with DEHP group. Quercetin 13-22 heme oxygenase 1 Homo sapiens 132-136 35024467-5 2022 Meanwhile, PEITC treatment ameliorated DON-induced an increase of the inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) mRNA levels and intracellular reactive oxygen species (ROS) level, and a decrease of glutathione peroxidase 1 (GPx1), superoxide dismutase 2 (SOD2), catalase (CAT) and heme oxygenase 1 (HO-1) mRNA levels. phenethyl isothiocyanate 11-16 heme oxygenase 1 Homo sapiens 324-328 35024467-5 2022 Meanwhile, PEITC treatment ameliorated DON-induced an increase of the inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) mRNA levels and intracellular reactive oxygen species (ROS) level, and a decrease of glutathione peroxidase 1 (GPx1), superoxide dismutase 2 (SOD2), catalase (CAT) and heme oxygenase 1 (HO-1) mRNA levels. deoxynivalenol 39-42 heme oxygenase 1 Homo sapiens 306-322 35024467-5 2022 Meanwhile, PEITC treatment ameliorated DON-induced an increase of the inducible nitric oxide synthase (iNOS) and cyclooxygenase 2 (COX-2) mRNA levels and intracellular reactive oxygen species (ROS) level, and a decrease of glutathione peroxidase 1 (GPx1), superoxide dismutase 2 (SOD2), catalase (CAT) and heme oxygenase 1 (HO-1) mRNA levels. deoxynivalenol 39-42 heme oxygenase 1 Homo sapiens 324-328 35045368-7 2022 Collectively, our studies demonstrated that compound D-6 protected against oxLDL-induced endothelial injury by activating Nrf2/HO-1 anti-oxidation pathway. oxynide 53-56 heme oxygenase 1 Homo sapiens 127-131 34999313-5 2022 In non-transfected cells, we also observed TNF-alpha-induced VCAM1 overexpression as well as heme-induced overexpression of HMOX1 in both cell models. Heme 93-97 heme oxygenase 1 Homo sapiens 124-129 35488673-0 2022 Raspberry ketone promotes FNDC5 protein expression via HO-1 upregulation in 3T3-L1 adipocytes. raspberry ketone 0-16 heme oxygenase 1 Homo sapiens 55-59 35085532-5 2022 Berberine had an antagonistic effect for the majority of genes mutual for AD and toxic metal mixture: ACHE, APP, BAX, BCL2, CASP3, HMOX1, IL1B, MAPT, SOD2, TNF. Berberine 0-9 heme oxygenase 1 Homo sapiens 131-136 35203060-0 2022 Metformin alleviates chronic obstructive pulmonary disease and cigarette smoke extract-induced glucocorticoid resistance by activating the nuclear factor E2-related factor 2/heme oxygenase-1 signaling pathway. Metformin 0-9 heme oxygenase 1 Homo sapiens 174-190 34973124-11 2022 The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. bardoxolone methyl 70-77 heme oxygenase 1 Homo sapiens 25-28 35218032-6 2022 Furthermore, preincubation with Orz reduced H2 O2 -mediated the proapoptotic protein of Bak expression and the phosphorylation of ASK1, p38, JNK, and ERK, and increased the anti-apoptotic protein of Bcl-xl expression and anti-oxidative stress proteins of Nrf2 and HO-1 expression. gamma-oryzanol 32-35 heme oxygenase 1 Homo sapiens 264-268 35123232-0 2022 Nicotinamide mononucleotide increases cell viability and restores tight junctions in high-glucose-treated human corneal epithelial cells via the SIRT1/Nrf2/HO-1 pathway. Nicotinamide Mononucleotide 0-27 heme oxygenase 1 Homo sapiens 156-160 35123232-0 2022 Nicotinamide mononucleotide increases cell viability and restores tight junctions in high-glucose-treated human corneal epithelial cells via the SIRT1/Nrf2/HO-1 pathway. Glucose 90-97 heme oxygenase 1 Homo sapiens 156-160 35268019-2 2022 ROS and inflammatory responses are regulated by the activation of nuclear factor erythroid-2-related factor 2 (Nrf2) and the expression of Nrf2 target genes, superoxide dismutase (SOD) and heme oxygenase-1 (HO-1). Reactive Oxygen Species 0-3 heme oxygenase 1 Homo sapiens 189-205 35268019-2 2022 ROS and inflammatory responses are regulated by the activation of nuclear factor erythroid-2-related factor 2 (Nrf2) and the expression of Nrf2 target genes, superoxide dismutase (SOD) and heme oxygenase-1 (HO-1). Reactive Oxygen Species 0-3 heme oxygenase 1 Homo sapiens 207-211 35268019-4 2022 We determined whether RGE suppresses the expression of IL-8 via Nrf2 activation and the expression of SOD and HO-1 in H. pylori-infected gastric epithelial AGS cells. (2s)-2-Amino-5-(2-(Methylsulfinyl)acetimidamido)pentanoic Acid 22-25 heme oxygenase 1 Homo sapiens 110-114 35268019-5 2022 H. pylori-infected cells were treated with RGE with or without ML385, an Nrf2 inhibitor, or zinc protoporphyrin (ZnPP), a HO-1 inhibitor. zinc protoporphyrin 92-111 heme oxygenase 1 Homo sapiens 122-126 35268019-5 2022 H. pylori-infected cells were treated with RGE with or without ML385, an Nrf2 inhibitor, or zinc protoporphyrin (ZnPP), a HO-1 inhibitor. zinc protoporphyrin 113-117 heme oxygenase 1 Homo sapiens 122-126 35268019-8 2022 H. pylori infection decreased the protein levels of SOD-1 and HO-1, as well as SOD activity, which was reversed by RGE treatment. (2s)-2-Amino-5-(2-(Methylsulfinyl)acetimidamido)pentanoic Acid 115-118 heme oxygenase 1 Homo sapiens 62-66 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Heme 110-114 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. ferrous iron 120-132 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Carbon Monoxide 134-149 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Carbon Monoxide 151-153 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Biliverdine 160-170 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Biliverdine 172-174 heme oxygenase 1 Homo sapiens 18-22 35048917-0 2022 Glucopyranose from Pleurotus geesteranus prevent alcoholic liver diseases by regulating Nrf2/HO-1-TLR4/NF-kappaB signalling pathways and gut microbiota. Glucose 0-13 heme oxygenase 1 Homo sapiens 93-97 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Bilirubin 176-185 heme oxygenase 1 Homo sapiens 18-22 35281042-2 2022 Heme oxygenase-1 (HO-1) is an essential stress-response enzyme highly expressed in the lungs, and catabolizes heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV)/bilirubin (BR), especially in pathological conditions which cause oxidative stress and inflammation. Bilirubin 187-189 heme oxygenase 1 Homo sapiens 18-22 35281042-7 2022 Being a highly infectious pathogenic bacterium, Mycobacterium tuberculosis (MTB) infection causes acute oxidative stress, and increases the expression of HO-1, which may in turn facilitate MTB survival and growth due to increased iron availability. Iron 230-234 heme oxygenase 1 Homo sapiens 154-158 35281042-8 2022 Moreover, in severe cases of MTB infection, excessive reactive oxygen species (ROS) and free iron (Fe2+) due to high levels of HO-1 can lead to lipid peroxidation and ferroptosis, which may promote further MTB dissemination from cells undergoing ferroptosis. Reactive Oxygen Species 54-77 heme oxygenase 1 Homo sapiens 127-131 35281042-8 2022 Moreover, in severe cases of MTB infection, excessive reactive oxygen species (ROS) and free iron (Fe2+) due to high levels of HO-1 can lead to lipid peroxidation and ferroptosis, which may promote further MTB dissemination from cells undergoing ferroptosis. Reactive Oxygen Species 79-82 heme oxygenase 1 Homo sapiens 127-131 35281042-8 2022 Moreover, in severe cases of MTB infection, excessive reactive oxygen species (ROS) and free iron (Fe2+) due to high levels of HO-1 can lead to lipid peroxidation and ferroptosis, which may promote further MTB dissemination from cells undergoing ferroptosis. Iron 93-97 heme oxygenase 1 Homo sapiens 127-131 35281042-8 2022 Moreover, in severe cases of MTB infection, excessive reactive oxygen species (ROS) and free iron (Fe2+) due to high levels of HO-1 can lead to lipid peroxidation and ferroptosis, which may promote further MTB dissemination from cells undergoing ferroptosis. ammonium ferrous sulfate 99-103 heme oxygenase 1 Homo sapiens 127-131 35218032-6 2022 Furthermore, preincubation with Orz reduced H2 O2 -mediated the proapoptotic protein of Bak expression and the phosphorylation of ASK1, p38, JNK, and ERK, and increased the anti-apoptotic protein of Bcl-xl expression and anti-oxidative stress proteins of Nrf2 and HO-1 expression. Hydrogen Peroxide 44-49 heme oxygenase 1 Homo sapiens 264-268 35204307-7 2022 We further found that 3,5,7-trimethoxyflavone suppressed the excessive increase in ROS, mitogen-activated protein kinases (MAPKs), Akt, and cyclooxygenase-2 (COX-2)and increased heme oxygenase (HO)-1 expression. 3,5,7-Trimethoxyflavone 22-45 heme oxygenase 1 Homo sapiens 178-199 35199318-8 2022 Both H2O2 and methemoglobin upregulated the expression of the HO-1 protein in the dTHP-1 monoculture; moreover, co-culture with endometriotic cells further enhanced HO-1 production. Hydrogen Peroxide 5-9 heme oxygenase 1 Homo sapiens 62-66 35199318-8 2022 Both H2O2 and methemoglobin upregulated the expression of the HO-1 protein in the dTHP-1 monoculture; moreover, co-culture with endometriotic cells further enhanced HO-1 production. Hydrogen Peroxide 5-9 heme oxygenase 1 Homo sapiens 165-169 35237380-0 2022 Ferritinophagy-Mediated Ferroptosis and Activation of Keap1/Nrf2/HO-1 Pathway Were Conducive to EMT Inhibition of Gastric Cancer Cells in Action of 2,2"-Di-pyridineketone Hydrazone Dithiocarbamate Butyric Acid Ester. 2,2"-di-pyridineketone hydrazone dithiocarbamate 148-196 heme oxygenase 1 Homo sapiens 65-69 35237380-0 2022 Ferritinophagy-Mediated Ferroptosis and Activation of Keap1/Nrf2/HO-1 Pathway Were Conducive to EMT Inhibition of Gastric Cancer Cells in Action of 2,2"-Di-pyridineketone Hydrazone Dithiocarbamate Butyric Acid Ester. butyric acid ester 197-215 heme oxygenase 1 Homo sapiens 65-69 35222803-8 2022 Silencing nuclear factor erythroid 2-related factor 2 (Nrf2) by targeted-siRNA and inhibiting heme oxygenase-1 (HO-1) through zinc protoporphyrin IX (ZnPP) significantly decreased cytoprotection of KGF-2. zinc protoporphyrin 126-148 heme oxygenase 1 Homo sapiens 94-110 35195252-5 2022 Specifically, FoxOs regulate mitochondrial biogenesis by dampening NRF1-Tfam and c-Myc-Tfam cascades directly, and inhibiting NAD-Sirt1-Pgc1alpha cascade indirectly by inducing Hmox1 or repressing Fxn and Urod. NAD 126-129 heme oxygenase 1 Homo sapiens 177-182 35222803-8 2022 Silencing nuclear factor erythroid 2-related factor 2 (Nrf2) by targeted-siRNA and inhibiting heme oxygenase-1 (HO-1) through zinc protoporphyrin IX (ZnPP) significantly decreased cytoprotection of KGF-2. zinc protoporphyrin 126-148 heme oxygenase 1 Homo sapiens 112-116 35222803-8 2022 Silencing nuclear factor erythroid 2-related factor 2 (Nrf2) by targeted-siRNA and inhibiting heme oxygenase-1 (HO-1) through zinc protoporphyrin IX (ZnPP) significantly decreased cytoprotection of KGF-2. zinc protoporphyrin 150-154 heme oxygenase 1 Homo sapiens 94-110 35222803-8 2022 Silencing nuclear factor erythroid 2-related factor 2 (Nrf2) by targeted-siRNA and inhibiting heme oxygenase-1 (HO-1) through zinc protoporphyrin IX (ZnPP) significantly decreased cytoprotection of KGF-2. zinc protoporphyrin 150-154 heme oxygenase 1 Homo sapiens 112-116 35222803-10 2022 As demonstrated by these data, KGF-2 resisted H2O2-mediated apoptosis and oxidative stress in HLECs through Nrf2/HO-1 and PI3K/Akt pathways, suggesting a potential protective effect against the formation of cataracts. Hydrogen Peroxide 46-50 heme oxygenase 1 Homo sapiens 113-117 35189631-10 2022 Furthermore, curcumin positively regulated the expression of Nrf2, HO-1, and SOD1 mRNAs in cells treated with 0.1 and 0.5 muM doxazosin. Curcumin 13-21 heme oxygenase 1 Homo sapiens 67-71 35209068-3 2022 In this study, we evaluated the ability of a topical liquid formula of polydeoxyribonucleotide (PDRN), vitamin C, and niacinamide (PVN) delivered via a microneedling therapy system (MTS) to attenuate photoaging and pigmentation by increasing nuclear factor erythroid 2-like 2 (NRF2)/heme oxygenase-1 (HO-1) and decreasing MMP expression in a UV-B-radiated animal model. pvn 131-134 heme oxygenase 1 Homo sapiens 283-299 35209068-3 2022 In this study, we evaluated the ability of a topical liquid formula of polydeoxyribonucleotide (PDRN), vitamin C, and niacinamide (PVN) delivered via a microneedling therapy system (MTS) to attenuate photoaging and pigmentation by increasing nuclear factor erythroid 2-like 2 (NRF2)/heme oxygenase-1 (HO-1) and decreasing MMP expression in a UV-B-radiated animal model. pvn 131-134 heme oxygenase 1 Homo sapiens 301-305 35189631-10 2022 Furthermore, curcumin positively regulated the expression of Nrf2, HO-1, and SOD1 mRNAs in cells treated with 0.1 and 0.5 muM doxazosin. Doxazosin 126-135 heme oxygenase 1 Homo sapiens 67-71 35197970-10 2022 Zinc sulfate significantly increased the levels of P-mTOR Ser2448, P-p70S6K Thr389, and P-tau Ser356 and decreased the levels of nuclear factor erythroid 2-related factor-2 (Nrf2) and heme oxygenase-1 (HO-1) in SH-SY5Y cells and in zinc-treated rats compared with the control groups. Zinc Sulfate 0-12 heme oxygenase 1 Homo sapiens 184-200 35144642-8 2022 Activated ROS-metabolism was identified in METTL7B-overexpressed LUAD cells, accompanied with upregulated protein level of GPX4, HMOX1 and SOD1 and their enzymatic activities. ros 10-13 heme oxygenase 1 Homo sapiens 129-134 35197970-10 2022 Zinc sulfate significantly increased the levels of P-mTOR Ser2448, P-p70S6K Thr389, and P-tau Ser356 and decreased the levels of nuclear factor erythroid 2-related factor-2 (Nrf2) and heme oxygenase-1 (HO-1) in SH-SY5Y cells and in zinc-treated rats compared with the control groups. Zinc Sulfate 0-12 heme oxygenase 1 Homo sapiens 202-206 34981336-10 2022 CONCLUSIONS: These findings suggest that WDR26 mediates H2O2-induced oxidative stress and cell injury, possibly by reducing the intrinsic apoptotic pathway and activating Nrf2 and HO-1 in astrocytes. Hydrogen Peroxide 56-60 heme oxygenase 1 Homo sapiens 180-184 35204177-9 2022 In addition, GBH restored the expression of ferroptosis marker proteins, such as GPX4, HO-1 and COX-2, which were altered by RSL3. gbh 13-16 heme oxygenase 1 Homo sapiens 87-91 34994954-8 2022 These phenomena, appeared to correlate with the recovered cellular antioxidant enzyme activities and inducted HO-1 (heme oxygenase-1) expression accompanying the nuclear translocation of Nrf2 conduct by saikosaponin-D preconditioning which had been altered by glutamate, were correlated with its neuroprotective. Glutamic Acid 260-269 heme oxygenase 1 Homo sapiens 110-114 34994954-8 2022 These phenomena, appeared to correlate with the recovered cellular antioxidant enzyme activities and inducted HO-1 (heme oxygenase-1) expression accompanying the nuclear translocation of Nrf2 conduct by saikosaponin-D preconditioning which had been altered by glutamate, were correlated with its neuroprotective. Glutamic Acid 260-269 heme oxygenase 1 Homo sapiens 116-132 34994954-11 2022 Taken together, our results indicated that saikosaponin-D enhanced cellular antioxidant capacity through not only intrinsic free radical-scavenging activity but also induction of endogenous antioxidant enzyme activities and HO-1 expression mediated, at least in part, by activating PI3K and subsequently Nrf2 nuclear translocation, thereby protecting the SH-SY5Y cells from glutamate-induced oxidative cytotoxicity. saikosaponin D 43-57 heme oxygenase 1 Homo sapiens 224-228 35204104-7 2022 Furthermore, the treatment of cells with HC-EA and its active compounds abolished intracellular ROS and increased levels of heme oxygenase-1 and superoxide dismutase. hc-ea 41-46 heme oxygenase 1 Homo sapiens 124-140 35204159-1 2022 Heme oxygenase 1 (HO-1), the rate-limiting enzyme in heme degradation, is involved in the maintenance of cellular homeostasis, exerting a cytoprotective role by its antioxidative and anti-inflammatory functions. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 35204159-1 2022 Heme oxygenase 1 (HO-1), the rate-limiting enzyme in heme degradation, is involved in the maintenance of cellular homeostasis, exerting a cytoprotective role by its antioxidative and anti-inflammatory functions. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 35204159-2 2022 HO-1 and its end products, biliverdin, carbon monoxide and free iron (Fe2+), confer cytoprotection against inflammatory and oxidative injury. Biliverdine 27-37 heme oxygenase 1 Homo sapiens 0-4 35204159-2 2022 HO-1 and its end products, biliverdin, carbon monoxide and free iron (Fe2+), confer cytoprotection against inflammatory and oxidative injury. Carbon Monoxide 39-54 heme oxygenase 1 Homo sapiens 0-4 35204159-2 2022 HO-1 and its end products, biliverdin, carbon monoxide and free iron (Fe2+), confer cytoprotection against inflammatory and oxidative injury. Iron 64-68 heme oxygenase 1 Homo sapiens 0-4 35204159-2 2022 HO-1 and its end products, biliverdin, carbon monoxide and free iron (Fe2+), confer cytoprotection against inflammatory and oxidative injury. ammonium ferrous sulfate 70-74 heme oxygenase 1 Homo sapiens 0-4 35043283-9 2022 Transcriptomic RNA-seq showed that FX abrogated TPA-induced differentially expressed genes (DEGs) of Nfe2l2-related genes Nqo1, Ho1, and Keap1. Tetradecanoylphorbol Acetate 48-51 heme oxygenase 1 Homo sapiens 128-131 35204062-3 2022 Exercise elevates heme oxygenase-1 (HO-1) and biliverdin reductase A (BVRA) expression as built-in protective mechanisms, which produce the most potent antioxidant, bilirubin. Bilirubin 165-174 heme oxygenase 1 Homo sapiens 18-34 35237424-8 2022 A 6 h pretreatment of 3-MA increased PM2.5-induced oxidative damage and cellular inflammation as indicated by increasing protein levels of HO-1, TXNIP, Bnip3L/NIX and IL-8 gene expression. 3-methyladenine 22-26 heme oxygenase 1 Homo sapiens 139-143 35204062-3 2022 Exercise elevates heme oxygenase-1 (HO-1) and biliverdin reductase A (BVRA) expression as built-in protective mechanisms, which produce the most potent antioxidant, bilirubin. Bilirubin 165-174 heme oxygenase 1 Homo sapiens 36-40 35204062-7 2022 The main objective of this review is to describe the function of bilirubin as an antioxidant and metabolic hormone and how the HO-1-BVRA-bilirubin-PPARalpha axis influences inflammation, metabolic function and interacts with exercise to improve outcomes of weight management. Bilirubin 137-146 heme oxygenase 1 Homo sapiens 127-131 35013107-9 2022 MET pretreatment also significantly suppressed MGO-stimulated ROS production, increased signaling through the ROS-mediated PI3K/Akt and Nrf2/HO-1 pathways, and markedly elevated the levels of its downstream antioxidants. Reactive Oxygen Species 110-113 heme oxygenase 1 Homo sapiens 141-145 35013107-12 2022 In addition, a PI3K inhibitor (LY-294002) and a Nrf2 inhibitor (ML385) observably attenuated the protective effects of MET on MGO-induced apoptosis and ROS generation by inhibiting the Nrf2/HO-1 pathways, while a ROS scavenger (NAC) and a permeability transition pores inhibitor (CsA) completely reversed these effects. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 31-40 heme oxygenase 1 Homo sapiens 190-194 35013107-12 2022 In addition, a PI3K inhibitor (LY-294002) and a Nrf2 inhibitor (ML385) observably attenuated the protective effects of MET on MGO-induced apoptosis and ROS generation by inhibiting the Nrf2/HO-1 pathways, while a ROS scavenger (NAC) and a permeability transition pores inhibitor (CsA) completely reversed these effects. ML385 64-69 heme oxygenase 1 Homo sapiens 190-194 35013107-12 2022 In addition, a PI3K inhibitor (LY-294002) and a Nrf2 inhibitor (ML385) observably attenuated the protective effects of MET on MGO-induced apoptosis and ROS generation by inhibiting the Nrf2/HO-1 pathways, while a ROS scavenger (NAC) and a permeability transition pores inhibitor (CsA) completely reversed these effects. Pyruvaldehyde 126-129 heme oxygenase 1 Homo sapiens 190-194 35047060-4 2022 Methods: An HO-1 inducer Hemin or an HO-1 inhibitor ZnPP-IX was used to treat the activated HSC-T6, respectively. Hemin 25-30 heme oxygenase 1 Homo sapiens 12-16 35047060-4 2022 Methods: An HO-1 inducer Hemin or an HO-1 inhibitor ZnPP-IX was used to treat the activated HSC-T6, respectively. zinc protoporphyrin 52-59 heme oxygenase 1 Homo sapiens 37-41 35056649-3 2022 In this work, we focused on determining the effect of kaempferol and its glycoside derivatives on the expression level of genes related to the reduction of oxidative stress-NFE2L2, NQO1, SOD1, SOD2, and HO-1; the enzymatic activity of superoxide dismutases; and the level of glutathione. kaempferol 54-64 heme oxygenase 1 Homo sapiens 203-207 35047556-0 2021 miR-3587 Inhibitor Attenuates Ferroptosis Following Renal Ischemia-Reperfusion Through HO-1. mir-3587 0-8 heme oxygenase 1 Homo sapiens 87-91 35047556-5 2021 When a miR-3587 inhibitor was applied in a hypoxia-reoxygenation (HR) model system using renal tubular epithelial cells, HO-1 protein (encoded by HMOX1) expression was significantly increased relative to that observed in the HR group, with concomitant increases in GPX4 protein levels, enhanced cell viability, a reduction in malondialdehyde content, decreased Fe2+ level, and the restoration of normal mitochondrial membrane potential. mir-3587 7-15 heme oxygenase 1 Homo sapiens 121-125 35047556-5 2021 When a miR-3587 inhibitor was applied in a hypoxia-reoxygenation (HR) model system using renal tubular epithelial cells, HO-1 protein (encoded by HMOX1) expression was significantly increased relative to that observed in the HR group, with concomitant increases in GPX4 protein levels, enhanced cell viability, a reduction in malondialdehyde content, decreased Fe2+ level, and the restoration of normal mitochondrial membrane potential. mir-3587 7-15 heme oxygenase 1 Homo sapiens 146-151 35047556-5 2021 When a miR-3587 inhibitor was applied in a hypoxia-reoxygenation (HR) model system using renal tubular epithelial cells, HO-1 protein (encoded by HMOX1) expression was significantly increased relative to that observed in the HR group, with concomitant increases in GPX4 protein levels, enhanced cell viability, a reduction in malondialdehyde content, decreased Fe2+ level, and the restoration of normal mitochondrial membrane potential. Malondialdehyde 326-341 heme oxygenase 1 Homo sapiens 121-125 35047556-5 2021 When a miR-3587 inhibitor was applied in a hypoxia-reoxygenation (HR) model system using renal tubular epithelial cells, HO-1 protein (encoded by HMOX1) expression was significantly increased relative to that observed in the HR group, with concomitant increases in GPX4 protein levels, enhanced cell viability, a reduction in malondialdehyde content, decreased Fe2+ level, and the restoration of normal mitochondrial membrane potential. ammonium ferrous sulfate 361-365 heme oxygenase 1 Homo sapiens 121-125 35047556-7 2021 Targeting of HMOX1 by miR-3587 was confirmed by luciferase reporter gene assay. mir-3587 22-30 heme oxygenase 1 Homo sapiens 13-18 35047556-8 2021 In conclusion, these preliminary results indicate that inhibition of miR-3587 promotes HO-1 upregulation, thereby protecting renal tissues from IR-induced ferroptosis. mir-3587 69-77 heme oxygenase 1 Homo sapiens 87-91 34986734-0 2022 Ramelteon protects against human pulmonary microvascular endothelial cell injury induced by lipopolysaccharide (LPS) via activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. ramelteon 0-9 heme oxygenase 1 Homo sapiens 183-199 34918393-5 2022 Our data indicate that overexpression of HO-1 or its metabolite biliverdin can maintain endothelial integrity upon DENV infection in vitro and in vivo. Biliverdine 64-74 heme oxygenase 1 Homo sapiens 41-45 34918393-7 2022 Subsequently, we confirmed that two medicinal plant-derived compounds, andrographolide, and celastrol, widely used as a nutritional or medicinal supplement are useful to attenuate DENV-induced plasma leakage through induction of the HO-1 expression in DENV-infected AG129 mice. andrographolide 71-86 heme oxygenase 1 Homo sapiens 233-237 34918393-7 2022 Subsequently, we confirmed that two medicinal plant-derived compounds, andrographolide, and celastrol, widely used as a nutritional or medicinal supplement are useful to attenuate DENV-induced plasma leakage through induction of the HO-1 expression in DENV-infected AG129 mice. celastrol 92-101 heme oxygenase 1 Homo sapiens 233-237 34986734-0 2022 Ramelteon protects against human pulmonary microvascular endothelial cell injury induced by lipopolysaccharide (LPS) via activating nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. ramelteon 0-9 heme oxygenase 1 Homo sapiens 201-205 34986734-12 2022 Ramelteon treatment also activated the Nrf2/HO-1 signaling pathway in LPS-induced HPMECs. ramelteon 0-9 heme oxygenase 1 Homo sapiens 44-48 34986734-14 2022 Collectively, the results suggested that ramelteon alleviated LPS-induced HPMEC damage by activating the Nrf2/HO-1 signaling pathway, making it an effective treatment for ALI. ramelteon 41-50 heme oxygenase 1 Homo sapiens 110-114 33982762-5 2021 Furthermore, THCA upregulated the expression levels of heme oxygenase-1 and NAD(P)H:quinone oxidoreductase 1, and the activation of nuclear factor erythroid 2-related factor 2 in HaCaT cells. thca 13-17 heme oxygenase 1 Homo sapiens 55-71 3290025-1 1988 In biological systems oxidation of heme is carried out by two isozymes of the microsomal heme oxygenase, HO-1 and HO-2. Heme 35-39 heme oxygenase 1 Homo sapiens 105-118 3290025-7 1988 Most likely, HO-1 gene is regulated in the same manner as metallothione in the gene. metallothione 58-71 heme oxygenase 1 Homo sapiens 13-17 3290025-9 1988 In vivo induction of HO-1 activity in the liver is accompanied by decreases in the total P-450 levels and, in a reconstituted system, cytochrome P-450b heme can be quantitatively converted to biliverdin by HO-1 and HO-2. Heme 152-156 heme oxygenase 1 Homo sapiens 21-25 3290025-9 1988 In vivo induction of HO-1 activity in the liver is accompanied by decreases in the total P-450 levels and, in a reconstituted system, cytochrome P-450b heme can be quantitatively converted to biliverdin by HO-1 and HO-2. Biliverdine 192-202 heme oxygenase 1 Homo sapiens 21-25 33862517-7 2021 The mechanism studies indicated that 13f exhibited cytoprotective effect by activating the Keap1-Nrf2-ARE pathway and stimulating downstream anti-oxidant protein including HO-1, NQO1, TrxR1, GCLC, and GCLM. 13F 37-40 heme oxygenase 1 Homo sapiens 172-176 479159-7 1979 The data indicate that there are regions of the hydrophobic disulfide knot, Ho1-DSK, which are surface-oriented. Disulfides 60-69 heme oxygenase 1 Homo sapiens 76-79 479159-8 1979 The distribution of iodine as mono- and diiodotyrosine in N-DSK and Ho1-DSK reflected the percentage (83 and 17, respectively) found in iodinated fibrinogen from which these fragments were prepared. Iodine 20-26 heme oxygenase 1 Homo sapiens 68-71 34025790-6 2021 Furthermore, it was demonstrated that the expression levels of the ferroptosis-associated genes heme oxygenase-1, glutamate-cysteine ligase catalytic and glutamate-cysteine ligase modifier were significantly upregulated after Andrographis treatment in both GC cell lines in reverse transcription-quantitative PCR experiments (P<0.05); this finding was further confirmed by immunoblotting assays (P<0.05). Glutamic Acid 114-123 heme oxygenase 1 Homo sapiens 96-112 34025790-6 2021 Furthermore, it was demonstrated that the expression levels of the ferroptosis-associated genes heme oxygenase-1, glutamate-cysteine ligase catalytic and glutamate-cysteine ligase modifier were significantly upregulated after Andrographis treatment in both GC cell lines in reverse transcription-quantitative PCR experiments (P<0.05); this finding was further confirmed by immunoblotting assays (P<0.05). Glutamic Acid 154-163 heme oxygenase 1 Homo sapiens 96-112 34025790-6 2021 Furthermore, it was demonstrated that the expression levels of the ferroptosis-associated genes heme oxygenase-1, glutamate-cysteine ligase catalytic and glutamate-cysteine ligase modifier were significantly upregulated after Andrographis treatment in both GC cell lines in reverse transcription-quantitative PCR experiments (P<0.05); this finding was further confirmed by immunoblotting assays (P<0.05). Cysteine 164-172 heme oxygenase 1 Homo sapiens 96-112 32712770-6 2021 In contrast, at 10 mumol.L-1 ziram and zinc associated-disulfiram induced expression of several antioxidants genes HMOX1, SOD2, and catalase, which could suggest the induction of oxidative stress. Ziram 29-34 heme oxygenase 1 Homo sapiens 115-120 32712770-6 2021 In contrast, at 10 mumol.L-1 ziram and zinc associated-disulfiram induced expression of several antioxidants genes HMOX1, SOD2, and catalase, which could suggest the induction of oxidative stress. Disulfiram 55-65 heme oxygenase 1 Homo sapiens 115-120 33905708-6 2021 RESULTS: Heme supports carcinogenesis via modulation of immune cell function, promoting inflammation and gut dysbiosis, impeding tumour suppressive potential of P53 gene, promoting cellular cytotoxicity and reactive oxygen species generation and modulating Nfr2 /HO-1 axis. Heme 9-13 heme oxygenase 1 Homo sapiens 263-267 33850557-0 2021 miR-125b/NRF2/HO-1 axis is involved in protection against oxidative stress of cystic fibrosis: A pilot study. mir-125b 0-8 heme oxygenase 1 Homo sapiens 14-18 34048148-0 2021 Lipoxin A4 protects primary spinal cord neurons from Erastin-induced ferroptosis by activating the Akt/Nrf2/HO-1 signaling pathway. lipoxin A4 0-10 heme oxygenase 1 Homo sapiens 108-112 33484436-14 2021 CONCLUSION: The precise mechanism accountable for the anti-inflammatory features of HO-1 is not completely understood; nevertheless, the CO signaling function associated with the antioxidant property shown by bilirubin possibly will play an act in the improvement of inflammation. Bilirubin 209-218 heme oxygenase 1 Homo sapiens 84-88 34048148-0 2021 Lipoxin A4 protects primary spinal cord neurons from Erastin-induced ferroptosis by activating the Akt/Nrf2/HO-1 signaling pathway. erastin 53-60 heme oxygenase 1 Homo sapiens 108-112 34048148-11 2021 In conclusion, this work demonstrated that LXA4 exerted a neuroprotective effect in Erastin-induced ferroptosis of primary spinal cord neurons by activating Akt/Nrf2/HO-1 signaling pathway. lipoxin A4 43-47 heme oxygenase 1 Homo sapiens 166-170 34048148-11 2021 In conclusion, this work demonstrated that LXA4 exerted a neuroprotective effect in Erastin-induced ferroptosis of primary spinal cord neurons by activating Akt/Nrf2/HO-1 signaling pathway. erastin 84-91 heme oxygenase 1 Homo sapiens 166-170 34049949-10 2021 Treatment of L2 and H441 cells with siRNAs against Nrf2 or HO-1 abrogated the protective effects of AICAR. AICA ribonucleotide 100-105 heme oxygenase 1 Homo sapiens 59-63 34012878-1 2021 AIM: To investigate the protective effect of heme oxygenase-1 (HO-1) against H2O2-induced apoptosis in human ARPE-19 cells. Hydrogen Peroxide 77-81 heme oxygenase 1 Homo sapiens 45-61 34049949-11 2021 CONCLUSIONS: Our data indicate that the primary mechanism for the protective action of AICAR in toxic gas injury is by upregulating lung HO-1 levels. AICA ribonucleotide 87-92 heme oxygenase 1 Homo sapiens 137-141 34039348-9 2021 Increased HO-1 further maintained reactive oxygen species (ROS) within a minimal tumor-promoting level and enhanced cytoprotective autophagy. Reactive Oxygen Species 34-57 heme oxygenase 1 Homo sapiens 10-14 34039348-9 2021 Increased HO-1 further maintained reactive oxygen species (ROS) within a minimal tumor-promoting level and enhanced cytoprotective autophagy. Reactive Oxygen Species 59-62 heme oxygenase 1 Homo sapiens 10-14 34012878-1 2021 AIM: To investigate the protective effect of heme oxygenase-1 (HO-1) against H2O2-induced apoptosis in human ARPE-19 cells. Hydrogen Peroxide 77-81 heme oxygenase 1 Homo sapiens 63-67 34012878-2 2021 METHODS: The lentiviral vector expressing HO-1 was prepared and transfected into apoptotic ARPE-19 cells induced by H2O2. Hydrogen Peroxide 116-120 heme oxygenase 1 Homo sapiens 42-46 34012878-5 2021 It was found that exogenous HO-1 significantly ameliorated H2O2-induced loss of cell viability, apoptosis and intracellular levels of reactive oxygen species (ROS) in ARPE-19 cells. Hydrogen Peroxide 59-63 heme oxygenase 1 Homo sapiens 28-32 34012878-5 2021 It was found that exogenous HO-1 significantly ameliorated H2O2-induced loss of cell viability, apoptosis and intracellular levels of reactive oxygen species (ROS) in ARPE-19 cells. Reactive Oxygen Species 134-157 heme oxygenase 1 Homo sapiens 28-32 34012878-5 2021 It was found that exogenous HO-1 significantly ameliorated H2O2-induced loss of cell viability, apoptosis and intracellular levels of reactive oxygen species (ROS) in ARPE-19 cells. Reactive Oxygen Species 159-162 heme oxygenase 1 Homo sapiens 28-32 34012878-7 2021 Furthermore, HO-1 upregulation further inhibited H2O2-induced release of cysteinyl aspartate specific proteinase-3 (caspase-3). Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 13-17 34012878-7 2021 Furthermore, HO-1 upregulation further inhibited H2O2-induced release of cysteinyl aspartate specific proteinase-3 (caspase-3). cysteinyl-aspartate 73-92 heme oxygenase 1 Homo sapiens 13-17 34012878-8 2021 CONCLUSION: Exogenous HO-1 protect ARPE-19 cells against H2O2-induced oxidative stress by regulating the expressions of Nrf2, HO-1, Bcl-2, and caspase-3. Hydrogen Peroxide 57-61 heme oxygenase 1 Homo sapiens 22-26 33999357-10 2021 Taken together, our findings suggest that activation of Nrf2/ARE/HO-1 signaling cascades contributes to PME-induced anti-oxidative and neuroprotective actions, which are at least partially mediated by AKT and ERK pathways. pinostrobin 104-107 heme oxygenase 1 Homo sapiens 65-69 33990765-3 2022 Preincubation with BELNs decreased the level of reactive oxygen species (ROS), increased the mitochondrial membrane potential, and prevented cell apoptosis by inducing the expression of Bcl-2 and heme oxygenase-1 (HO-1) and decreasing the content of Bax in rotenone-treated HepG2 cells. belns 19-24 heme oxygenase 1 Homo sapiens 196-212 34011367-0 2021 Esomeprazole attenuates inflammatory and fibrotic response in lung cells through the MAPK/Nrf2/HO1 pathway. Esomeprazole 0-12 heme oxygenase 1 Homo sapiens 95-98 34011367-6 2021 RESULTS: Our study shows that esomeprazole controls pro-inflammatory and profibrotic molecules through nuclear translocation of the transcription factor nuclear factor-like 2 (Nrf2) and induction of the cytoprotective molecule heme oxygenase 1 (HO1). Esomeprazole 30-42 heme oxygenase 1 Homo sapiens 227-243 34011367-6 2021 RESULTS: Our study shows that esomeprazole controls pro-inflammatory and profibrotic molecules through nuclear translocation of the transcription factor nuclear factor-like 2 (Nrf2) and induction of the cytoprotective molecule heme oxygenase 1 (HO1). Esomeprazole 30-42 heme oxygenase 1 Homo sapiens 245-248 34011367-7 2021 Genetic deletion of Nrf2 or pharmacological inhibition of HO1 impaired esomeprazole-mediated regulation of proinflammatory and profibrotic molecules. Esomeprazole 71-83 heme oxygenase 1 Homo sapiens 58-61 33990765-3 2022 Preincubation with BELNs decreased the level of reactive oxygen species (ROS), increased the mitochondrial membrane potential, and prevented cell apoptosis by inducing the expression of Bcl-2 and heme oxygenase-1 (HO-1) and decreasing the content of Bax in rotenone-treated HepG2 cells. belns 19-24 heme oxygenase 1 Homo sapiens 214-218 33903766-2 2021 Here we show that a distinct subset of TAMs (F4/80hiCD115hiC3aRhiCD88hi), endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), plays a critical role in shaping a prometastatic tumor microenvironment favoring immunosuppression, angiogenesis and epithelial-to-mesenchymal transition. Heme 101-105 heme oxygenase 1 Homo sapiens 149-165 34025070-16 2021 Moreover, SB203580 significantly reduced the pulmonary P-p38, NFkappaB, P-signal transducer and activator of transcription-3 and Myd88 levels but increased the IkappaB and HO-1 levels. SB 203580 10-18 heme oxygenase 1 Homo sapiens 172-176 33845576-0 2021 A Coumarin-Porphyrin FRET Break-Apart Probe for Heme Oxygenase-1. coumarin 2-10 heme oxygenase 1 Homo sapiens 48-64 33845576-1 2021 Heme oxygenase-1 (HO-1) is a vital enzyme in humans that primarily regulates free heme concentrations. Heme 82-86 heme oxygenase 1 Homo sapiens 0-16 33845576-1 2021 Heme oxygenase-1 (HO-1) is a vital enzyme in humans that primarily regulates free heme concentrations. Heme 82-86 heme oxygenase 1 Homo sapiens 18-22 33845576-4 2021 Reported here are the design, synthesis, and photophysical and biological characterization of a coumarin-porphyrin FRET break-apart probe to detect HO-1 activity, Fe-L1. coumarin 96-104 heme oxygenase 1 Homo sapiens 148-152 33845576-5 2021 We designed Fe-L1 to "break-apart" upon HO-1-catalyzed porphyrin degradation, perturbing the efficient FRET mechanism from a coumarin donor to a porphyrin acceptor fluorophore. Porphyrins 55-64 heme oxygenase 1 Homo sapiens 40-44 33845576-5 2021 We designed Fe-L1 to "break-apart" upon HO-1-catalyzed porphyrin degradation, perturbing the efficient FRET mechanism from a coumarin donor to a porphyrin acceptor fluorophore. coumarin 125-133 heme oxygenase 1 Homo sapiens 40-44 33845576-5 2021 We designed Fe-L1 to "break-apart" upon HO-1-catalyzed porphyrin degradation, perturbing the efficient FRET mechanism from a coumarin donor to a porphyrin acceptor fluorophore. Porphyrins 145-154 heme oxygenase 1 Homo sapiens 40-44 33845576-6 2021 Analysis of HO-1 activity using Escherichia coli lysates overexpressing hHO-1 found that a 6-fold increase in emission intensity at 383 nm was observed following incubation with NADPH. NADP 178-183 heme oxygenase 1 Homo sapiens 12-16 33845576-6 2021 Analysis of HO-1 activity using Escherichia coli lysates overexpressing hHO-1 found that a 6-fold increase in emission intensity at 383 nm was observed following incubation with NADPH. NADP 178-183 heme oxygenase 1 Homo sapiens 72-77 33845576-8 2021 Finally, through the analysis of Fe-L2, we have shown that close structural analogues of heme are required to maintain HO-1 activity. Heme 89-93 heme oxygenase 1 Homo sapiens 119-123 34017261-0 2021 Xanthohumol Attenuated Inflammation and ECM Degradation by Mediating HO-1/C/EBPbeta Pathway in Osteoarthritis Chondrocytes. xanthohumol 0-11 heme oxygenase 1 Homo sapiens 69-73 34017261-10 2021 HO-1 knockdown could abrogate the protective effects of XH in IL-1beta-treated chondrocytes. xanthohumol 56-58 heme oxygenase 1 Homo sapiens 0-4 33388910-1 2021 Heme oxygenase (HO)-1 is a rate-limiting enzyme for degrading heme into carbon monoxide. Heme 62-66 heme oxygenase 1 Homo sapiens 0-21 33388910-1 2021 Heme oxygenase (HO)-1 is a rate-limiting enzyme for degrading heme into carbon monoxide. Carbon Monoxide 72-87 heme oxygenase 1 Homo sapiens 0-21 33617922-0 2021 Dihydroactinidiolide regulates Nrf2/HO-1 expression and inhibits caspase-3/Bax pathway to protect SH-SY5Y human neuroblastoma cells from oxidative stress induced neuronal apoptosis. dihydroactinidiolide 0-20 heme oxygenase 1 Homo sapiens 36-40 33617922-6 2021 Western blotting and RT-PCR revealed that DHAc significantly increased anti-apoptotic Bcl-2 expression and mRNA levels of Nrf2 and HO-1. dihydroactinidiolide 42-46 heme oxygenase 1 Homo sapiens 131-135 33980059-0 2021 Curcumin Can Activate the Nrf2/HO-1 Signaling Pathway and Scavenge Free Radicals in Spinal Cord Injury Treatment. Curcumin 0-8 heme oxygenase 1 Homo sapiens 31-35 33980059-6 2021 In this review, we analyze the role of curcumin in activating Nrf2/HO-1 and scavenging free radicals to repair SCI. Curcumin 39-47 heme oxygenase 1 Homo sapiens 67-71 33977953-6 2021 In oleic acid (OA)-induced HepG2 cells, hesperetin upregulated antioxidant levels (SOD/GPx/GR/GCLC/HO-1) by triggering the PI3 K/AKT-Nrf2 pathway, alleviating OA-induced reactive oxygen species (ROS) overproduction and hepatotoxicity. hesperetin 40-50 heme oxygenase 1 Homo sapiens 99-103 33955073-4 2021 These recent studies have demonstrated that carotenoids also provide photoprotection against UVA-induced pigmentation and inhibit molecular markers of oxidative stress such as intercellular adhesion molecule-1, heme-oxygenase-1, matrix metalloproteinase-1, 9, and others. Carotenoids 44-55 heme oxygenase 1 Homo sapiens 211-227 34055200-0 2021 Corrigendum to "Myricetin Possesses Potential Protective Effects on Diabetic Cardiomyopathy through Inhibiting IkappaBalpha/NFkappaB and Enhancing Nrf2/HO-1". myricetin 16-25 heme oxygenase 1 Homo sapiens 152-156 32893669-0 2021 Cpt1a promoted ROS-induced oxidative stress and inflammation in liver injury via the Nrf2/HO-1 and NLRP3 inflammasome signaling pathway. Reactive Oxygen Species 15-18 heme oxygenase 1 Homo sapiens 90-94 33903766-2 2021 Here we show that a distinct subset of TAMs (F4/80hiCD115hiC3aRhiCD88hi), endowed with high rates of heme catabolism by the stress-responsive enzyme heme oxygenase-1 (HO-1), plays a critical role in shaping a prometastatic tumor microenvironment favoring immunosuppression, angiogenesis and epithelial-to-mesenchymal transition. Heme 101-105 heme oxygenase 1 Homo sapiens 167-171 33744612-12 2021 Baicalin relieved the above changes caused by DHAV-1 and activated the gene and protein expressions of Nrf2, which activated ARE-dependent genes including heme oxygenase-1 (HO-1), nicotinamide adenine dinucleotide phosphate quinone oxidoreductase 1 (NQO1), SOD-1, and GPX-1. baicalin 0-8 heme oxygenase 1 Homo sapiens 155-171 33744612-12 2021 Baicalin relieved the above changes caused by DHAV-1 and activated the gene and protein expressions of Nrf2, which activated ARE-dependent genes including heme oxygenase-1 (HO-1), nicotinamide adenine dinucleotide phosphate quinone oxidoreductase 1 (NQO1), SOD-1, and GPX-1. baicalin 0-8 heme oxygenase 1 Homo sapiens 173-177 33986683-8 2021 Furthermore, the inhibitor of the enzyme heme oxygenase-1 but not that of nitric oxide synthase reduced GUO-induced neurite outgrowth. Guanosine 104-107 heme oxygenase 1 Homo sapiens 41-57 33975215-0 2021 Associations between blood heavy metal(loid)s and serum heme oxygenase-1 in pregnant women: Do their distribution patterns matter? Metals 33-38 heme oxygenase 1 Homo sapiens 56-72 33981228-7 2021 Meanwhile, galangin increased ROS accumulation, and reduced Nrf2 and NQO-1, but elevated HO-1 in MGC 803 cells. galangin 11-19 heme oxygenase 1 Homo sapiens 89-93 33902245-7 2021 Additionally, the concentration of PAHs was negatively correlated with the level of HO-1 (r=-0.358, P=0.017). Polycyclic Aromatic Hydrocarbons 35-39 heme oxygenase 1 Homo sapiens 84-88 33902245-8 2021 Linear regression analysis showed that the concentration of HO-1 decreased by 1.737 mug/L when the concentration of PAHs increased by 1 mg/L (P=0.035). Polycyclic Aromatic Hydrocarbons 116-120 heme oxygenase 1 Homo sapiens 60-64 33922144-5 2021 Here, we investigated the protective effects of THF on neurotoxicity induced by METH exposure by enhancing the Nrf2/HO-1 and PI3K/Akt/mTOR signaling pathways in SH-SY5y cells. Methamphetamine 80-84 heme oxygenase 1 Homo sapiens 116-120 33922144-7 2021 Moreover, we found THF induced Nrf2 nuclear translocation and HO-1 expression. tetrahydrofuran 19-22 heme oxygenase 1 Homo sapiens 62-66 33922144-5 2021 Here, we investigated the protective effects of THF on neurotoxicity induced by METH exposure by enhancing the Nrf2/HO-1 and PI3K/Akt/mTOR signaling pathways in SH-SY5y cells. tetrahydrofuran 48-51 heme oxygenase 1 Homo sapiens 116-120 33922144-8 2021 An inhibitor assay confirmed that the induction of HO-1 by THF attenuates METH-induced neurotoxicity. tetrahydrofuran 59-62 heme oxygenase 1 Homo sapiens 51-55 33922144-8 2021 An inhibitor assay confirmed that the induction of HO-1 by THF attenuates METH-induced neurotoxicity. Methamphetamine 74-78 heme oxygenase 1 Homo sapiens 51-55 33922144-9 2021 Therefore, we suggest that THF preserves neuronal cells from METH-induced neurotoxicity by upregulating HO-1 expression through the Nrf2 and PI3K/Akt/mTOR signaling pathways. tetrahydrofuran 27-30 heme oxygenase 1 Homo sapiens 104-108 33919311-4 2021 The results of voxel-based multiple regression analysis showed that blood levels of CypA, HO-1, and IRE1 were correlated with GMV on brain magnetic resonance imaging (MRI) in the entire population (p = 0.0005). Guanosine-5'-Phosphovanadate 126-129 heme oxygenase 1 Homo sapiens 90-94 33919311-7 2021 HO-1 values increased with increasing GMV at the uncus (r = 0.307, p = 0.0008), lateral globus pallidus and putamen (r = 0.287, p = 0.002), and hippocampus (r = 0.197, p = 0.034). Guanosine-5'-Phosphovanadate 38-41 heme oxygenase 1 Homo sapiens 0-4 33924157-0 2021 (E)-Guggulsterone Inhibits Dengue Virus Replication by Upregulating Antiviral Interferon Responses through the Induction of Heme Oxygenase-1 Expression. pregna-4,17-diene-3,16-dione 0-17 heme oxygenase 1 Homo sapiens 124-140 33923922-8 2021 These were effectively antagonized with MGN (20 muM), which led to HUVECs being protected against DOX-induced apoptosis, partly through the PI3K/AKT-mediated NRF2/HO-1 signaling pathway, which could theoretically protect the vessels from severe DOX toxicity. Doxorubicin 98-101 heme oxygenase 1 Homo sapiens 163-167 33924157-9 2021 The mechanistic studied revealed that (E)-guggulsterone stimulates Nrf2-mediated heme oxygenase-1 (HO-1) expression, which increases the antiviral interferon responses and downstream antiviral gene expression by blocking DENV NS2B/3B protease activity. pregna-4,17-diene-3,16-dione 38-55 heme oxygenase 1 Homo sapiens 81-97 33924157-9 2021 The mechanistic studied revealed that (E)-guggulsterone stimulates Nrf2-mediated heme oxygenase-1 (HO-1) expression, which increases the antiviral interferon responses and downstream antiviral gene expression by blocking DENV NS2B/3B protease activity. pregna-4,17-diene-3,16-dione 38-55 heme oxygenase 1 Homo sapiens 99-103 33994055-9 2021 CONCLUSION: These results indicate that AU can reduce inflammation and oxidative stress through the NF-kappaB, Nrf2/HO-1, and MAPK pathways. aucubin 40-42 heme oxygenase 1 Homo sapiens 116-120 33875681-3 2021 Here, we observed that curcumin induced the expression of genes downstream of Nrf2 such as HO-1, NQO1, and GST-mu1 in neuronal cells, and increased the level of Nrf2 protein. Curcumin 23-31 heme oxygenase 1 Homo sapiens 91-95 33854358-0 2021 Natural Dietary Supplement, Carvacrol, Alleviates LPS-Induced Oxidative Stress, Neurodegeneration, and Depressive-Like Behaviors via the Nrf2/HO-1 Pathway. carvacrol 28-37 heme oxygenase 1 Homo sapiens 142-146 33923585-0 2021 Preventive Effects against Retinal Degeneration by Centella asiatica Extract (CA-HE50) and Asiaticoside through Apoptosis Suppression by the Nrf2/HO-1 Signaling Pathway. ca-he50 78-85 heme oxygenase 1 Homo sapiens 146-150 33923585-0 2021 Preventive Effects against Retinal Degeneration by Centella asiatica Extract (CA-HE50) and Asiaticoside through Apoptosis Suppression by the Nrf2/HO-1 Signaling Pathway. asiaticoside 91-103 heme oxygenase 1 Homo sapiens 146-150 33920891-0 2021 Heme-Mediated Activation of the Nrf2/HO-1 Axis Attenuates Calcification of Valve Interstitial Cells. Heme 0-4 heme oxygenase 1 Homo sapiens 37-41 33920891-7 2021 Heme induced Nrf2 and HO-1 expression in VICs. Heme 0-4 heme oxygenase 1 Homo sapiens 22-26 33920891-8 2021 Heme lost its anti-calcification potential when we blocked transcriptional activity Nrf2 or enzyme activity of HO-1. Heme 0-4 heme oxygenase 1 Homo sapiens 111-115 33920891-10 2021 This study suggests that heme-mediated activation of the Nrf2/HO-1 pathway inhibits the calcification of VICs. Heme 25-29 heme oxygenase 1 Homo sapiens 62-66 33678013-14 2022 The GI safety of ATB-352 could be due to the involvement of heme oxygenase-1 and SOCS3 pathways activation. atb-352 17-24 heme oxygenase 1 Homo sapiens 60-76 33927798-7 2021 Additionally, in clinical patients who received laparoscopic gynecological surgery, pretreatment with hydromorphone resulted in lower serum levels of club cell secretory protein-16 (CC-16) and intercellular adhesion molecule-1 (ICAM-1), a lower prooxidant-antioxidant balance (PAB), and higher heme oxygenase-1 (HO-1) activity than morphine pretreatment. Hydromorphone 102-115 heme oxygenase 1 Homo sapiens 294-310 33927798-7 2021 Additionally, in clinical patients who received laparoscopic gynecological surgery, pretreatment with hydromorphone resulted in lower serum levels of club cell secretory protein-16 (CC-16) and intercellular adhesion molecule-1 (ICAM-1), a lower prooxidant-antioxidant balance (PAB), and higher heme oxygenase-1 (HO-1) activity than morphine pretreatment. Hydromorphone 102-115 heme oxygenase 1 Homo sapiens 312-316 33927798-8 2021 Collectively, our results suggest that hydromorphone protects against CO2 pneumoperitoneum-induced lung injury via HO-1-regulated mitochondrial dynamics and may be a promising strategy to treat CO2 pneumoperitoneum-induced lung injury. Hydromorphone 39-52 heme oxygenase 1 Homo sapiens 115-119 33927798-8 2021 Collectively, our results suggest that hydromorphone protects against CO2 pneumoperitoneum-induced lung injury via HO-1-regulated mitochondrial dynamics and may be a promising strategy to treat CO2 pneumoperitoneum-induced lung injury. Carbon Dioxide 194-197 heme oxygenase 1 Homo sapiens 115-119 33923744-3 2021 The contribution of HO-1 in redox homeostasis leads to a relevant decrease in cells oxidative damage, which can be reconducted to its cytoprotective effects explicated alongside other endogenous mechanisms involving genes like TIGAR (TP53-induced glycolysis and apoptosis regulator), but also to the therapeutic functions of heme main transformation products, especially carbon monoxide (CO), which has been shown to be effective on GSH levels implementation sustaining body"s antioxidant response to oxidative stress. Heme 325-329 heme oxygenase 1 Homo sapiens 20-24 33923744-3 2021 The contribution of HO-1 in redox homeostasis leads to a relevant decrease in cells oxidative damage, which can be reconducted to its cytoprotective effects explicated alongside other endogenous mechanisms involving genes like TIGAR (TP53-induced glycolysis and apoptosis regulator), but also to the therapeutic functions of heme main transformation products, especially carbon monoxide (CO), which has been shown to be effective on GSH levels implementation sustaining body"s antioxidant response to oxidative stress. Carbon Monoxide 371-386 heme oxygenase 1 Homo sapiens 20-24 33923744-3 2021 The contribution of HO-1 in redox homeostasis leads to a relevant decrease in cells oxidative damage, which can be reconducted to its cytoprotective effects explicated alongside other endogenous mechanisms involving genes like TIGAR (TP53-induced glycolysis and apoptosis regulator), but also to the therapeutic functions of heme main transformation products, especially carbon monoxide (CO), which has been shown to be effective on GSH levels implementation sustaining body"s antioxidant response to oxidative stress. Carbon Monoxide 388-390 heme oxygenase 1 Homo sapiens 20-24 33923744-3 2021 The contribution of HO-1 in redox homeostasis leads to a relevant decrease in cells oxidative damage, which can be reconducted to its cytoprotective effects explicated alongside other endogenous mechanisms involving genes like TIGAR (TP53-induced glycolysis and apoptosis regulator), but also to the therapeutic functions of heme main transformation products, especially carbon monoxide (CO), which has been shown to be effective on GSH levels implementation sustaining body"s antioxidant response to oxidative stress. Glutathione 433-436 heme oxygenase 1 Homo sapiens 20-24 33860917-7 2021 Furthermore, activation of Nrf-2/HO-1 signaling and upregulation of HSP-70 governs the protection elicited by daphnetin against oxidative stress-induced neuronal apoptosis. daphnetin 110-119 heme oxygenase 1 Homo sapiens 33-37 33863580-8 2021 In endometriosis, total iron showed a positive correlation with HO-1 (r, 0.518, p = 0.001), but there were no antioxidants that correlated with iron in non-endometriosis. Iron 24-28 heme oxygenase 1 Homo sapiens 64-68 33863580-10 2021 CONCLUSIONS: HO-1 may regulate the delicate balance of iron-induced oxidative stress in endometriotic cyst fluid. Iron 55-59 heme oxygenase 1 Homo sapiens 13-17 33854363-8 2021 In further analysis, we found that hub bridge genes HLA-A, HMOX1, and JUN related to endocytosis, cell adhesion, and phagosomes may exert their effects through the dopamine (DA) system and the serotonergic pathway post-ICH depression. Dopamine 164-172 heme oxygenase 1 Homo sapiens 59-64 33917842-12 2021 EGCG increased antioxidant levels (HO-1) at week-4 (p < 0.01) plus elastin at week-8 (p < 0.01). epigallocatechin gallate 0-4 heme oxygenase 1 Homo sapiens 35-39 33854363-8 2021 In further analysis, we found that hub bridge genes HLA-A, HMOX1, and JUN related to endocytosis, cell adhesion, and phagosomes may exert their effects through the dopamine (DA) system and the serotonergic pathway post-ICH depression. Dopamine 174-176 heme oxygenase 1 Homo sapiens 59-64 33754715-1 2021 Human heme oxygenase (hHO-1) is a physiologically important enzyme responsible for free heme catabolism. Heme 6-10 heme oxygenase 1 Homo sapiens 22-27 33754715-9 2021 These studies suggest that the FeCN fragment in hHO-1 is tilted more strongly toward the porphyrin macrocycle compared to other histidine-ligated proteins, reflecting the propensity of the exogenous hHO-l ligands to position toward the alpha-meso-carbon, which is crucial for the HO reactivity and essential for regioselectivity. Histidine 128-137 heme oxygenase 1 Homo sapiens 48-53 33754715-9 2021 These studies suggest that the FeCN fragment in hHO-1 is tilted more strongly toward the porphyrin macrocycle compared to other histidine-ligated proteins, reflecting the propensity of the exogenous hHO-l ligands to position toward the alpha-meso-carbon, which is crucial for the HO reactivity and essential for regioselectivity. alpha-meso-carbon 236-253 heme oxygenase 1 Homo sapiens 48-53 33831425-0 2021 Cephalosporin antibiotics specifically and selectively target nasopharyngeal carcinoma through HMOX1-induced ferroptosis. Cephalosporins 0-13 heme oxygenase 1 Homo sapiens 95-100 33831425-8 2021 Mechanistic assessment of HMOX1 in cefotaxime-mediated ferroptosis was tested with Protoporphyrin IX zinc. Cefotaxime 35-45 heme oxygenase 1 Homo sapiens 26-31 33831425-8 2021 Mechanistic assessment of HMOX1 in cefotaxime-mediated ferroptosis was tested with Protoporphyrin IX zinc. protoporphyrin IX 83-100 heme oxygenase 1 Homo sapiens 26-31 33831425-13 2021 Inhibition of HMOX1 significantly reduced the anticancer efficacy of cefotaxime in CNE2 cells. Cefotaxime 69-79 heme oxygenase 1 Homo sapiens 14-19 33831425-15 2021 We mechanistically show that induction of ferroptosis through HMOX1 induction mediates cefotaxime anticancer activity. Cefotaxime 87-97 heme oxygenase 1 Homo sapiens 62-67 33393728-5 2021 In addition, mangiferin attenuated the enhancement of p-JNK, reductions of Nrf2 and HO-1, and increased level of the mitochondrial fission proteins Drp1 caused by fluoride. mangiferin 13-23 heme oxygenase 1 Homo sapiens 84-88 33898397-8 2021 The mechanism experiments showed that the antitumor activity of ORI-NPs against breast cancer might be through ROS related Nrf2/HO-1 signaling pathway. ros 111-114 heme oxygenase 1 Homo sapiens 128-132 33898501-1 2021 Fubaiju Hot-Water Extracts Against ARPE-19 Cell Oxidative Damage by Activating PI3K/Akt-Mediated Nrf2/HO-1 Signaling Pathway. fubaiju 0-7 heme oxygenase 1 Homo sapiens 102-106 33898501-1 2021 Fubaiju Hot-Water Extracts Against ARPE-19 Cell Oxidative Damage by Activating PI3K/Akt-Mediated Nrf2/HO-1 Signaling Pathway. Water 12-17 heme oxygenase 1 Homo sapiens 102-106 33898501-8 2021 Meanwhile, CM treatment upregulated Akt phosphorylation, nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, and the expression level of antioxidant gene heme oxygenase-1 (HO-1) in a dose-dependent manner under oxidative stress. Curium 11-13 heme oxygenase 1 Homo sapiens 194-198 33453607-14 2021 HMOX1 knockdown attenuated EF24-induced cytotoxicity and attenuated EF24-induced inhibition of Glutathione Peroxidase 4 (GPX4) expression. EF24 27-31 heme oxygenase 1 Homo sapiens 0-5 33453607-14 2021 HMOX1 knockdown attenuated EF24-induced cytotoxicity and attenuated EF24-induced inhibition of Glutathione Peroxidase 4 (GPX4) expression. EF24 68-72 heme oxygenase 1 Homo sapiens 0-5 33453607-15 2021 CONCLUSION: Our results showed that EF24 upregulated HMOX1 to suppress GPX4 expression to induce ferroptosis by increasing MDA level, ROS level and intracellular ferric ion level. Reactive Oxygen Species 134-137 heme oxygenase 1 Homo sapiens 53-58 33453607-16 2021 Thus, EF24 might serve as a potential agent for the treatment of HMOX1-positive osteosarcoma patients. EF24 6-10 heme oxygenase 1 Homo sapiens 65-70 33270331-9 2021 Notably, the expression of antioxidant genes, such as HO-1, and the nuclear translocation of Nrf2 were further enhanced by F-FEJS in UVB/H2 O2 -treated cells. Hydrogen Peroxide 137-142 heme oxygenase 1 Homo sapiens 54-58 33898501-10 2021 Correspondingly, the protective effect of CM was dramatically blocked after interference with phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor LY294002, indicating that the protective effect of CM on cell oxidative damage was attributed to PI3K/Akt-mediated Nrf2/HO-1 signaling pathway. Curium 42-44 heme oxygenase 1 Homo sapiens 265-269 33898501-10 2021 Correspondingly, the protective effect of CM was dramatically blocked after interference with phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor LY294002, indicating that the protective effect of CM on cell oxidative damage was attributed to PI3K/Akt-mediated Nrf2/HO-1 signaling pathway. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 145-153 heme oxygenase 1 Homo sapiens 265-269 33898501-10 2021 Correspondingly, the protective effect of CM was dramatically blocked after interference with phosphatidylinositol 3-kinase (PI3K)/Akt inhibitor LY294002, indicating that the protective effect of CM on cell oxidative damage was attributed to PI3K/Akt-mediated Nrf2/HO-1 signaling pathway. Curium 196-198 heme oxygenase 1 Homo sapiens 265-269 33987332-9 2021 Conclusions: Our work suggests that the Nrf2/HO-1 molecular pathway may play a crucial role in treating ICH patients with PGE1 and may represent novel molecular targets, resulting in discovering new drugs for ICH treatment. Alprostadil 122-126 heme oxygenase 1 Homo sapiens 45-49 33636332-7 2021 Taken all together, we demonstrate that plasma treated water-derived oxidants sensitize pancreatic cancer cells to ferroptotic cell death by targeting a NRF2-HMOX1-GPX4 specific kinase signaling network. Water 55-60 heme oxygenase 1 Homo sapiens 158-163 33393728-7 2021 In conclusion, protection by mangiferin against fluoride-induced neurotoxicity involves normalizing the impaired mitochondrial apoptotic pathway and dynamics and reducing oxidative stress via inactivation of the JNK and activation of the Nrf2/HO-1 pathways. mangiferin 29-39 heme oxygenase 1 Homo sapiens 243-247 33393728-7 2021 In conclusion, protection by mangiferin against fluoride-induced neurotoxicity involves normalizing the impaired mitochondrial apoptotic pathway and dynamics and reducing oxidative stress via inactivation of the JNK and activation of the Nrf2/HO-1 pathways. Fluorides 48-56 heme oxygenase 1 Homo sapiens 243-247 33537805-2 2021 HO-1 can be induced by a variety of stimuli, including heavy metals, heat shock, inflammatory stimuli, heme and its derivatives, stress, hypoxia, and biological hormones. Heme 103-107 heme oxygenase 1 Homo sapiens 0-4 33448057-8 2021 CsA-induced oxidative stress, HO-1, TGF-beta1, and type II EMT were also rescued by antioxidants treatment. Cyclosporine 0-3 heme oxygenase 1 Homo sapiens 30-34 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Heme 36-40 heme oxygenase 1 Homo sapiens 0-4 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Heme 66-70 heme oxygenase 1 Homo sapiens 0-4 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Biliverdine 76-86 heme oxygenase 1 Homo sapiens 0-4 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 0-4 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Carbon Monoxide 105-107 heme oxygenase 1 Homo sapiens 0-4 33537805-3 2021 HO-1 is the rate-limiting enzyme of heme catabolism, which splits heme into biliverdin, carbon monoxide (CO) and iron. Iron 113-117 heme oxygenase 1 Homo sapiens 0-4 33537805-7 2021 In particular, this review focuses on the role of carbon monoxide (CO), one of the primary metabolites of HO-1, in flap survival and discusses the feasibility and existing challenges of HO-1 in flap surgery. Carbon Monoxide 50-65 heme oxygenase 1 Homo sapiens 106-110 33537805-7 2021 In particular, this review focuses on the role of carbon monoxide (CO), one of the primary metabolites of HO-1, in flap survival and discusses the feasibility and existing challenges of HO-1 in flap surgery. Carbon Monoxide 67-69 heme oxygenase 1 Homo sapiens 106-110 33513620-4 2021 To understand the underlying mechanism, we investigated the effect of low-dose TSA on HO-1, SOD and CAT induction and activating Akt together with its downstream Nrf2 signaling pathway. trichostatin A 79-82 heme oxygenase 1 Homo sapiens 86-90 32930996-10 2021 Inhibition of heme oxygenase-1 (HO-1) also blocked the EM-induced mitochondrial protection. Emodin 55-57 heme oxygenase 1 Homo sapiens 14-30 32930996-10 2021 Inhibition of heme oxygenase-1 (HO-1) also blocked the EM-induced mitochondrial protection. Emodin 55-57 heme oxygenase 1 Homo sapiens 32-36 32930996-11 2021 Therefore, EM protected the mitochondria by a mechanism dependent on the AMPK/Nrf2/HO-1 signaling pathway in MG-challenged SH-SY5Y cells. Pyruvaldehyde 109-111 heme oxygenase 1 Homo sapiens 83-87 33915898-8 2021 Intriguingly, MiR-100 antagomir treatment exerted a cytoprotective effect against the H2O2-induced oxidative cell death through attenuating the oxidation-induced AMPK hyperphosphorylation, restoring cellular mTOR and p62/SQSTM1 levels and upregulating heme oxygenase-1 expression. Hydrogen Peroxide 86-90 heme oxygenase 1 Homo sapiens 252-268 33646477-0 2021 East to West not North-West: Structure-Based Mechanistic Resolution of 8-Hydroxyl Replacement and Resulting Effects on the Activities of Imidazole-Based Heme Oxygenase-1 Inhibitors. imidazole 137-146 heme oxygenase 1 Homo sapiens 153-169 33646477-3 2021 In a recent study, two selective azole-based HO-1 inhibitors (Cpd1 and Cpd2) were synthesized, which exhibited differential inhibitory potencies of ~200mum. Azoles 33-38 heme oxygenase 1 Homo sapiens 45-49 33646477-6 2021 Findings revealed favorable binding for the bromobenzene and imidazole substituents of Cpd1 at the western and eastern regions of the HO-1 active domain. bromobenzene 44-56 heme oxygenase 1 Homo sapiens 134-138 33646477-6 2021 Findings revealed favorable binding for the bromobenzene and imidazole substituents of Cpd1 at the western and eastern regions of the HO-1 active domain. imidazole 61-70 heme oxygenase 1 Homo sapiens 134-138 33513620-7 2021 Therefore, our results suggest that low-dose TSA can activate the Akt/Nrf2 pathway and upregulate expression of HO-1, SOD and CAT to stimulate the cellular defense mechanism. trichostatin A 45-48 heme oxygenase 1 Homo sapiens 112-116 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. Heme 73-77 heme oxygenase 1 Homo sapiens 91-95 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. Biliverdine 131-141 heme oxygenase 1 Homo sapiens 73-89 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. Biliverdine 131-141 heme oxygenase 1 Homo sapiens 91-95 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. ammonium ferrous sulfate 143-154 heme oxygenase 1 Homo sapiens 73-89 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. ammonium ferrous sulfate 143-154 heme oxygenase 1 Homo sapiens 91-95 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. Carbon Monoxide 159-174 heme oxygenase 1 Homo sapiens 73-89 33787980-3 2021 In this review, we summarize how this complex system is regulated by the heme oxygenase-1 (HO-1)-an enzyme, which degrades heme to biliverdin, ferrous ion and carbon monoxide. Carbon Monoxide 159-174 heme oxygenase 1 Homo sapiens 91-95 33787980-8 2021 We critically discuss the issue of metalloporphyrins, commonly used pharmacological modulators of HO-1 activity, and raise the issue of their important HO-1-independent activities. Metalloporphyrins 35-52 heme oxygenase 1 Homo sapiens 98-102 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Heme 126-130 heme oxygenase 1 Homo sapiens 0-16 33781184-0 2021 Micro Composite Palmitoylethanolamide/Rutin Reduces Vascular Injury through Modulation of the Nrf2/HO-1 and NF-kB Pathways. palmidrol 16-37 heme oxygenase 1 Homo sapiens 99-103 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Heme 126-130 heme oxygenase 1 Homo sapiens 18-22 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Carbon Monoxide 184-199 heme oxygenase 1 Homo sapiens 0-16 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Carbon Monoxide 184-199 heme oxygenase 1 Homo sapiens 18-22 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Carbon Monoxide 201-203 heme oxygenase 1 Homo sapiens 0-16 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Carbon Monoxide 201-203 heme oxygenase 1 Homo sapiens 18-22 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Biliverdine 206-216 heme oxygenase 1 Homo sapiens 0-16 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). Biliverdine 206-216 heme oxygenase 1 Homo sapiens 18-22 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). ferrous iron 221-233 heme oxygenase 1 Homo sapiens 0-16 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). ferrous iron 221-233 heme oxygenase 1 Homo sapiens 18-22 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). ammonium ferrous sulfate 235-239 heme oxygenase 1 Homo sapiens 0-16 33868304-1 2021 Heme oxygenase-1 (HO-1) is an inducible intracellular enzyme that is expressed in response to a variety of stimuli to degrade heme, which generates the biologically active catabolites carbon monoxide (CO), biliverdin and ferrous iron (Fe2+). ammonium ferrous sulfate 235-239 heme oxygenase 1 Homo sapiens 18-22 33868304-4 2021 Intrinsically to the cell, HO-1 activity provides antioxidant, anti-apoptotic and cytoprotective effects via its catabolites as well as clearing toxic intracellular heme. Heme 165-169 heme oxygenase 1 Homo sapiens 27-31 33781184-0 2021 Micro Composite Palmitoylethanolamide/Rutin Reduces Vascular Injury through Modulation of the Nrf2/HO-1 and NF-kB Pathways. Rutin 38-43 heme oxygenase 1 Homo sapiens 99-103 33625409-12 2021 d-pinitol not only significantly activated the Nrf2/ARE signaling pathway via facilitating the translocation of Nrf2 from sitoplazma to cytoblast, upregulating the protein expression levels of GCLC, GCLM, HO-1, and NQO1, but also improved the PPAR-gamma level by binding to the active site of PPAR-gamma, when suppressing NF-kappaB p65 and IkappaBalpha phosphorylation. pinitol 0-9 heme oxygenase 1 Homo sapiens 205-209 33547170-11 2021 Inhibition of Asah2 increased p53 protein stability to upregulate Hmox1 expression to suppress lipid reactive oxygen species production to suppress ferroptosis in MDSCs. lipid reactive oxygen species 95-124 heme oxygenase 1 Homo sapiens 66-71 33453713-0 2021 MiR-133a-3p relieves the oxidative stress induced trophoblast cell apoptosis through the BACH1/Nrf2/HO-1 signaling pathway. mir-133a-3p 0-11 heme oxygenase 1 Homo sapiens 100-104 33453713-12 2021 It is collectively demonstrated that miR-133a-3p can relieve the oxidative stress-induced apoptosis in the trophoblast cells through the BACH1/Nrf2/HO-1 signaling pathway via targeting BACH1 directly. mir-133a 37-45 heme oxygenase 1 Homo sapiens 148-152 33738869-8 2021 Heme oxygenase-1 (HO-1), a scavenger of ROS, was triggered by HIF-1 alpha. Reactive Oxygen Species 40-43 heme oxygenase 1 Homo sapiens 0-16 33738869-8 2021 Heme oxygenase-1 (HO-1), a scavenger of ROS, was triggered by HIF-1 alpha. Reactive Oxygen Species 40-43 heme oxygenase 1 Homo sapiens 18-22 33738869-9 2021 The effect of hydrogen peroxide was inhibited by HO-1 induction, whereas it was promoted by HO-1 knockdown. Hydrogen Peroxide 14-31 heme oxygenase 1 Homo sapiens 49-53 33738869-10 2021 HO-1 inhibition by zinc protoporphyrin promoted the differentiation and increased ROS. zinc protoporphyrin 19-38 heme oxygenase 1 Homo sapiens 0-4 33738869-10 2021 HO-1 inhibition by zinc protoporphyrin promoted the differentiation and increased ROS. Reactive Oxygen Species 82-85 heme oxygenase 1 Homo sapiens 0-4 33738869-12 2021 Immature HL cells are inhibited from differentiation by a reduction of ROS through the induction of HO-1 via HIF-1alpha. Reactive Oxygen Species 71-74 heme oxygenase 1 Homo sapiens 100-104 33733282-16 2021 CONCLUSIONS: A certain concentration range of CSPE can increase the expression of the downstream protein HO-1 and negatively regulate the production of ROS by upregulating the expression of Nrf2, thus protecting human retinal endothelial cells from oxidative damage caused by high glucose. Glucose 281-288 heme oxygenase 1 Homo sapiens 105-109 33189376-7 2021 Likewise, serum Cu was positively associated with HO-1 (beta = 0.0004, P = 0.03) and negatively associated with MCP-1 (beta = -0.0006, P = 0.003) and IL-8 (beta = -0.002, P = 0.03). Copper 16-18 heme oxygenase 1 Homo sapiens 50-54 33465425-8 2021 In addition, brusatol significantly suppressed the expression of Nrf2 and HO-1, and abrogated tBHQ-induced Nrf2 activation. brusatol 13-21 heme oxygenase 1 Homo sapiens 74-78 33577831-0 2021 Ergothioneine alleviates senescence of fibroblasts induced by UVB damage of keratinocytes via activation of the Nrf2/HO-1 pathway and HSP70 in keratinocytes. Ergothioneine 0-13 heme oxygenase 1 Homo sapiens 117-121 33566044-8 2021 Furthermore, the stimulating effects of PS-ALA on the PGC-1alpha/Nrf1/Tfam pathway and Nrf2/HO-1 pathway as well as its mitochondrial biogenesis promotion effects and anti-oxidative activities were abrogated by the AMPK inhibitor in OA-treated HepG2 cells. ps-ala 40-46 heme oxygenase 1 Homo sapiens 92-96 33491566-3 2021 HIF1 pathway inhibitor digoxin blocked hypoxia-stimulated HIF1alpha activation and heme oxygenase (HMOX1) expression in HK2 cells. Digoxin 23-30 heme oxygenase 1 Homo sapiens 99-104 33491566-4 2021 Hypoxia mimicking CoCl2 stimulated HMOX1 expression was significantly lower in Nrf2-/- RTECs than in WT counterparts. cobaltous chloride 18-23 heme oxygenase 1 Homo sapiens 35-40 32782233-7 2021 Antioxidant response element (ARE)-reporter activity and expression of ARE-dependent genes NAD(P)H dehydrogenase [quinone] 1 (NQO1), heme oxygenase-1 (HO-1) were increased by resorcinol. resorcinol 175-185 heme oxygenase 1 Homo sapiens 133-149 32782233-7 2021 Antioxidant response element (ARE)-reporter activity and expression of ARE-dependent genes NAD(P)H dehydrogenase [quinone] 1 (NQO1), heme oxygenase-1 (HO-1) were increased by resorcinol. resorcinol 175-185 heme oxygenase 1 Homo sapiens 151-155 33245515-9 2021 RT-qPCR showed that GA significantly increased the mRNA levels of Heme oxygenase-1 (HO-1) and NADPH:quinone oxidoreductase1 (NQO1). Gallium 20-22 heme oxygenase 1 Homo sapiens 66-82 33245515-9 2021 RT-qPCR showed that GA significantly increased the mRNA levels of Heme oxygenase-1 (HO-1) and NADPH:quinone oxidoreductase1 (NQO1). Gallium 20-22 heme oxygenase 1 Homo sapiens 84-88 33571414-6 2021 Real-time qPCR quantification and Western blotting verified that the GA pretreatment elevates NAD(P)H quinone oxidoreductase-1 (NQO1) by more than 4.6-fold, heme oxygenase (HO-1) by about 1.2-fold, and the glutamate-cysteine ligase catalytic (GCLC) subunit by 0.7-fold. ginnalin A 69-71 heme oxygenase 1 Homo sapiens 173-177 33678318-6 2021 RESULTS: In ectopic endometrial cells, caffeic acid caused a significant elevation in Nrf-2 gene expression level, NQO1, and HO-1 enzyme activities. caffeic acid 39-51 heme oxygenase 1 Homo sapiens 125-129 33388347-5 2021 Sulforaphane treatment induced the activity of acetaldehyde-metabolizing mitochondrial aldehyde dehydrogenase (ALDH2) in HepaRG cells and suppressed the acetaldehyde-induced proliferation and profibrogenic activity in LX-2 cells with upregulation of Nrf2-regulated antioxidant genes, including HMOX1, NQO1, and GSTM3. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 294-299 33388347-5 2021 Sulforaphane treatment induced the activity of acetaldehyde-metabolizing mitochondrial aldehyde dehydrogenase (ALDH2) in HepaRG cells and suppressed the acetaldehyde-induced proliferation and profibrogenic activity in LX-2 cells with upregulation of Nrf2-regulated antioxidant genes, including HMOX1, NQO1, and GSTM3. Acetaldehyde 47-59 heme oxygenase 1 Homo sapiens 294-299 33058354-10 2021 Indeed, cynaropicrin increased the expression of several antioxidant genes, such as glutamate-cysteine ligase and heme oxygenase-1, by promoting the activation of the transcription factor Nrf-2. cynaropicrin 8-20 heme oxygenase 1 Homo sapiens 114-130 33664571-0 2021 Copper Nanoparticles Induce Oxidative Stress via the Heme Oxygenase 1 Signaling Pathway in vitro Studies. Copper 0-6 heme oxygenase 1 Homo sapiens 53-69 33647480-9 2021 Emodin treatment activated the Nuclear factorery throid 2-related factor 2/Kelch-like ECH-associated protein 1-antioxidatant response element (Nrf2/Keap1-ARE) pathway and upregulated the expression of heme oxygenase 1 (HO-1), superoxide dismutase (SOD), and catalase (CAT) in the hepatopancreas after CyHV-3 infection. Emodin 0-6 heme oxygenase 1 Homo sapiens 201-217 33647480-9 2021 Emodin treatment activated the Nuclear factorery throid 2-related factor 2/Kelch-like ECH-associated protein 1-antioxidatant response element (Nrf2/Keap1-ARE) pathway and upregulated the expression of heme oxygenase 1 (HO-1), superoxide dismutase (SOD), and catalase (CAT) in the hepatopancreas after CyHV-3 infection. Emodin 0-6 heme oxygenase 1 Homo sapiens 219-223 33671004-8 2021 In contrast, in ferroptosis, HO-1 may play a pro-death role via enhancing iron release. Iron 74-78 heme oxygenase 1 Homo sapiens 29-33 33716792-6 2021 Heme oxygenase-1 (HO-1), a phase II detoxification enzyme responsible for heme catabolism, is induced by oxidative stress, among others. Heme 74-78 heme oxygenase 1 Homo sapiens 0-16 33664571-6 2021 Subsequently, for oxidative stress parameter detection, the cells were pre-treated with hemin to induce HO-1 expression prior to CuNP treatment. Hemin 88-93 heme oxygenase 1 Homo sapiens 104-108 33716792-6 2021 Heme oxygenase-1 (HO-1), a phase II detoxification enzyme responsible for heme catabolism, is induced by oxidative stress, among others. Heme 74-78 heme oxygenase 1 Homo sapiens 18-22 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. Heme 23-27 heme oxygenase 1 Homo sapiens 208-212 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. Oxides 141-146 heme oxygenase 1 Homo sapiens 208-212 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. Carbon Monoxide 354-369 heme oxygenase 1 Homo sapiens 208-212 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. Biliverdine 371-381 heme oxygenase 1 Homo sapiens 208-212 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. Bilirubin 382-391 heme oxygenase 1 Homo sapiens 208-212 33716792-8 2021 The regulatory role of heme availability for the synthesis of enzymes involved in hypertension development, such as cyclooxygenase or nitric oxide synthase, seems to be responsible for many of the beneficial HO-1 effects; additionally, the antioxidant, anti-inflammatory, antiapoptotic, and antiproliferative effects of the end products of its reaction, carbon monoxide, biliverdin/bilirubin, and Fe2+, would also contribute. ammonium ferrous sulfate 397-401 heme oxygenase 1 Homo sapiens 208-212 33664571-8 2021 Furthermore, hemin pretreatment induced HO-1 expression in cells, which partially reduced the accumulation of reactive oxygen species induced by CuNPs and increased the levels of antioxidant enzymes. Hemin 13-18 heme oxygenase 1 Homo sapiens 40-44 33664571-8 2021 Furthermore, hemin pretreatment induced HO-1 expression in cells, which partially reduced the accumulation of reactive oxygen species induced by CuNPs and increased the levels of antioxidant enzymes. Oxygen 119-125 heme oxygenase 1 Homo sapiens 40-44 33664571-8 2021 Furthermore, hemin pretreatment induced HO-1 expression in cells, which partially reduced the accumulation of reactive oxygen species induced by CuNPs and increased the levels of antioxidant enzymes. cunps 145-150 heme oxygenase 1 Homo sapiens 40-44 33579280-0 2021 AITC inhibits fibroblast-myofibroblast transition via TRPA1-independent MAPK and NRF2/HO-1 pathways and reverses corticosteroids insensitivity in human lung fibroblasts. allyl isothiocyanate 0-4 heme oxygenase 1 Homo sapiens 86-90 33316526-0 2021 Activation of the ROS/HO-1/NQO1 signaling pathway contributes to the copper-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Reactive Oxygen Species 18-21 heme oxygenase 1 Homo sapiens 22-26 33316526-0 2021 Activation of the ROS/HO-1/NQO1 signaling pathway contributes to the copper-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Copper 69-75 heme oxygenase 1 Homo sapiens 22-26 33316526-1 2021 The aim of this study was to investigate the crosstalk between oxidative stress and autophagy through the ROS/HO-1/NQO1 pathway caused by copper (Cu). Reactive Oxygen Species 106-109 heme oxygenase 1 Homo sapiens 110-114 33316526-1 2021 The aim of this study was to investigate the crosstalk between oxidative stress and autophagy through the ROS/HO-1/NQO1 pathway caused by copper (Cu). Copper 138-144 heme oxygenase 1 Homo sapiens 110-114 33316526-1 2021 The aim of this study was to investigate the crosstalk between oxidative stress and autophagy through the ROS/HO-1/NQO1 pathway caused by copper (Cu). Copper 146-148 heme oxygenase 1 Homo sapiens 110-114 33316526-5 2021 In summary, our findings indicated that excessive Cu could induce oxidative stress and autophagy by activating the ROS/HO-1/NQO1 pathway, and inhibition of HO-1 might attenuate Cu-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Copper 50-52 heme oxygenase 1 Homo sapiens 119-123 33316526-5 2021 In summary, our findings indicated that excessive Cu could induce oxidative stress and autophagy by activating the ROS/HO-1/NQO1 pathway, and inhibition of HO-1 might attenuate Cu-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Reactive Oxygen Species 115-118 heme oxygenase 1 Homo sapiens 119-123 33316526-5 2021 In summary, our findings indicated that excessive Cu could induce oxidative stress and autophagy by activating the ROS/HO-1/NQO1 pathway, and inhibition of HO-1 might attenuate Cu-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Copper 177-179 heme oxygenase 1 Homo sapiens 119-123 33316526-5 2021 In summary, our findings indicated that excessive Cu could induce oxidative stress and autophagy by activating the ROS/HO-1/NQO1 pathway, and inhibition of HO-1 might attenuate Cu-induced oxidative stress and autophagy in duck renal tubular epithelial cells. Copper 177-179 heme oxygenase 1 Homo sapiens 156-160 33643023-2 2021 Biliverdin is derived from heme through a reaction mediated by HO-1 and protects cells from oxidative stress. Biliverdine 0-10 heme oxygenase 1 Homo sapiens 63-67 33643023-2 2021 Biliverdin is derived from heme through a reaction mediated by HO-1 and protects cells from oxidative stress. Heme 27-31 heme oxygenase 1 Homo sapiens 63-67 33643023-3 2021 However, iron and carbon monoxide produced by the catabolism of HO-1 exert detrimental effects on patients with PD. Iron 9-13 heme oxygenase 1 Homo sapiens 64-68 33643023-3 2021 However, iron and carbon monoxide produced by the catabolism of HO-1 exert detrimental effects on patients with PD. Carbon Monoxide 18-33 heme oxygenase 1 Homo sapiens 64-68 33669070-4 2021 Biochemical studies showed that Zn(II)-curc treatment increased the NRF2 protein levels along with its targets, heme oxygenase-1 (HO-1) and p62/SQSTM1, while markedly reduced the levels of Keap1 (Kelch-like ECH-associated protein 1), the NRF2 inhibitor, in the cancer cell lines analyzed. zn(ii) 32-38 heme oxygenase 1 Homo sapiens 112-128 33669070-4 2021 Biochemical studies showed that Zn(II)-curc treatment increased the NRF2 protein levels along with its targets, heme oxygenase-1 (HO-1) and p62/SQSTM1, while markedly reduced the levels of Keap1 (Kelch-like ECH-associated protein 1), the NRF2 inhibitor, in the cancer cell lines analyzed. zn(ii) 32-38 heme oxygenase 1 Homo sapiens 130-134 33668397-0 2021 Protective Effects of 6,7,4"-Trihydroxyflavanone on Hypoxia-Induced Neurotoxicity by Enhancement of HO-1 through Nrf2 Signaling Pathway. 6,7,4'-Trihydroxyflavanone 22-48 heme oxygenase 1 Homo sapiens 100-104 33668397-7 2021 We revealed that treatment with THF promotes HO-1 expression through Nrf2 nuclear translocation. tetrahydrofuran 32-35 heme oxygenase 1 Homo sapiens 45-49 33668397-8 2021 An inhibitor assay using tin protoporphyrin IX (SnPP) confirmed that the enhancement of HO-1 by pre-treatment with THF protects SH-SY5y cells from CoCl2-induced neurotoxicity under hypoxic conditions. tin protoporphyrin IX 25-46 heme oxygenase 1 Homo sapiens 88-92 33668397-8 2021 An inhibitor assay using tin protoporphyrin IX (SnPP) confirmed that the enhancement of HO-1 by pre-treatment with THF protects SH-SY5y cells from CoCl2-induced neurotoxicity under hypoxic conditions. S-Nitroso-N-propionyl-D,L-penicillamine 48-52 heme oxygenase 1 Homo sapiens 88-92 33668397-8 2021 An inhibitor assay using tin protoporphyrin IX (SnPP) confirmed that the enhancement of HO-1 by pre-treatment with THF protects SH-SY5y cells from CoCl2-induced neurotoxicity under hypoxic conditions. tetrahydrofuran 115-118 heme oxygenase 1 Homo sapiens 88-92 33668397-8 2021 An inhibitor assay using tin protoporphyrin IX (SnPP) confirmed that the enhancement of HO-1 by pre-treatment with THF protects SH-SY5y cells from CoCl2-induced neurotoxicity under hypoxic conditions. cobaltous chloride 147-152 heme oxygenase 1 Homo sapiens 88-92 33668397-9 2021 Our results demonstrate the advantageous effects of THF against hypoxia-induced neurotoxicity through the HO-1/Nrf2 signaling pathway and provide a therapeutic insight for neurodegenerative disorders. tetrahydrofuran 52-55 heme oxygenase 1 Homo sapiens 106-110 33579280-8 2021 AITC significantly increased phosphorylated- extracellular signal-regulated kinase (ERK) 1/2 and heme oxygenase (HO)-1 protein expressions (P < 0.05) in TGF-beta1-treated cells. allyl isothiocyanate 0-4 heme oxygenase 1 Homo sapiens 97-118 33579280-12 2021 CONCLUSION: We found that AITC exerts protective effect on TGF-beta1-induced alpha-SMA induction by activating ERK1/2 MAPK and NRF2/HO-1 pathways in lung fibroblasts. allyl isothiocyanate 26-30 heme oxygenase 1 Homo sapiens 132-136 33581965-9 2021 Results from qRT-PCR indicated that the suppression of Nrf2 and HO-1 following AGEs stimulation was reversed by Magnolol. magnolol 112-120 heme oxygenase 1 Homo sapiens 64-68 33546257-7 2021 Two peptides and their combination treatment resulted in the increased heme oxygenase-1 (HO-1), a phase II detoxifying enzyme, through the activation of nuclear transcription factor-erythroid 2-related factor (Nrf2). Peptides 4-12 heme oxygenase 1 Homo sapiens 89-93 33546433-6 2021 Differential array analysis revealed early modulation of stress response gene expression in both A375 melanoma and PANC-1 adenocarcinoma cells elicited by D2O (90%; <=6 h) (upregulated: CDKN1A, DDIT3, EGR1, GADD45A, HMOX1, NFKBIA, or SOD2 (up to 9-fold; p < 0.01)) confirmed by independent RT-qPCR analysis. Deuterium Oxide 155-158 heme oxygenase 1 Homo sapiens 216-221 33546257-7 2021 Two peptides and their combination treatment resulted in the increased heme oxygenase-1 (HO-1), a phase II detoxifying enzyme, through the activation of nuclear transcription factor-erythroid 2-related factor (Nrf2). Peptides 4-12 heme oxygenase 1 Homo sapiens 71-87 33569875-5 2021 Green tea catechins especially epigallocatechin gallate (EGCG) elucidated the protective role of Nrf2 and its downstream molecules in various disorders through Keap-1, HO-1, NQO-1, GPx, GCLc, GCLm, NF-kB cross-link, kinases, and apoptotic proteins. Catechin 10-19 heme oxygenase 1 Homo sapiens 168-172 33569875-5 2021 Green tea catechins especially epigallocatechin gallate (EGCG) elucidated the protective role of Nrf2 and its downstream molecules in various disorders through Keap-1, HO-1, NQO-1, GPx, GCLc, GCLm, NF-kB cross-link, kinases, and apoptotic proteins. epigallocatechin gallate 31-55 heme oxygenase 1 Homo sapiens 168-172 33569875-5 2021 Green tea catechins especially epigallocatechin gallate (EGCG) elucidated the protective role of Nrf2 and its downstream molecules in various disorders through Keap-1, HO-1, NQO-1, GPx, GCLc, GCLm, NF-kB cross-link, kinases, and apoptotic proteins. epigallocatechin gallate 57-61 heme oxygenase 1 Homo sapiens 168-172 33547785-12 2022 Two thirds of intervention studies (50% dietary, such as using resistant starch) reported an increase of NRF2, NQO1, or HO-1. Starch 73-79 heme oxygenase 1 Homo sapiens 120-124 33603823-5 2021 Mechanistic studies revealed that the protective effects of GFE were related to its activation of Nrf2 and HO-1 signaling in HaCaT cells. 1-(3,4-dichlorophenyl)-6,6-dimethyl-1,3,5-triazine-2,4-diamine 60-63 heme oxygenase 1 Homo sapiens 107-111 33347928-7 2021 Besides, polysaccharides show protective effects through certain signaling pathways including PI3K/Akt, MAPK, and NF-kappaB, PARP-1/AIF, JNK3/c-Jun/Fas-L, and Nrf2/HO-1 signaling pathways. Polysaccharides 9-24 heme oxygenase 1 Homo sapiens 164-168 32372266-8 2021 Resveratrol directly upregulates antioxidative capacity by increasing antioxidant genes such as heme oxygenase-1, superoxide dismutase, catalase, and glutathione. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 96-112 33645502-13 2021 Introduction of high concentration of fisetin promoted the translocation of Nrf2 from the cytoplasm into the nucleus, increasing protein expression of its downstream elements, at least HO-1 in IR liver tissues and hepatocytes after HR. fisetin 38-45 heme oxygenase 1 Homo sapiens 185-189 33645502-15 2021 CONCLUSION: Fisetin alleviates liver damage, cell apoptosis, and oxidative stress induced by liver IRI, at least through Nrf2/HO-1 signaling pathway, suggesting that fisetin could be considered as a targeted drug for liver IRI treatment. fisetin 12-19 heme oxygenase 1 Homo sapiens 126-130 33645502-15 2021 CONCLUSION: Fisetin alleviates liver damage, cell apoptosis, and oxidative stress induced by liver IRI, at least through Nrf2/HO-1 signaling pathway, suggesting that fisetin could be considered as a targeted drug for liver IRI treatment. fisetin 166-173 heme oxygenase 1 Homo sapiens 126-130 33023966-8 2021 This increase in HO-1 protein was recapitulated in human A375 melanoma cells exposed to fructose in culture. Fructose 88-96 heme oxygenase 1 Homo sapiens 17-21 33287596-6 2021 Heme oxygenase-1 (HO-1) induction offers protection via inhibition of NADPH oxidase and promotion of cGMP generation. Cyclic GMP 101-105 heme oxygenase 1 Homo sapiens 0-16 33287596-6 2021 Heme oxygenase-1 (HO-1) induction offers protection via inhibition of NADPH oxidase and promotion of cGMP generation. Cyclic GMP 101-105 heme oxygenase 1 Homo sapiens 18-22 33310139-7 2021 Garcinone D (GarD) significantly upregulated AhR/Cyp1a1 and Nrf2/HO-1 protein expression and enhanced zonula occludens-1 and occludin protein levels in HT-29 cells. garcinone 0-9 heme oxygenase 1 Homo sapiens 65-69 32964457-13 2021 The inhibitory effect on neuronal damage is achieved through the antioxidant (via inducing the Nrf2/HO-1 system) and anti-inflammatory effects (via promoting JNK/AP-1 signaling) of DHA. Docosahexaenoic Acids 181-184 heme oxygenase 1 Homo sapiens 100-104 33355378-4 2021 However, whether 0.7% ISO affords protection against septic neuronal injury involving HO-1 activation is unclear. iso 22-25 heme oxygenase 1 Homo sapiens 86-90 33176981-0 2021 Desaturation and heme elevation during COVID-19 infection: A potential prognostic factor of heme oxygenase-1. Heme 17-21 heme oxygenase 1 Homo sapiens 92-108 33573363-5 2021 The obtained results showed that PPJE significantly inhibited reactive oxygen species release and increased the cellular antioxidant response, as indicated by the increased expression of cytoprotective enzymes, such as heme oxygenase-1 and NAD(P)H dehydrogenase [quinone] 1. ppje 33-37 heme oxygenase 1 Homo sapiens 219-235 33514947-4 2021 In this Review, we present an introduction to HO-1 for immunologists, including an overview of its roles in iron metabolism and antioxidant defence, and the factors which regulate its expression. Iron 108-112 heme oxygenase 1 Homo sapiens 46-50 33512783-12 2021 The cells and the wound tissues treated with BB showed highest Nrf2 activation and HO-1 expression than PVP-I and CHG. BB was highly efficient in wound infection control while delayed wound healing. boeravinone B 45-47 heme oxygenase 1 Homo sapiens 83-87 33512783-12 2021 The cells and the wound tissues treated with BB showed highest Nrf2 activation and HO-1 expression than PVP-I and CHG. BB was highly efficient in wound infection control while delayed wound healing. boeravinone B 119-121 heme oxygenase 1 Homo sapiens 83-87 33394027-9 2021 This interaction between CYP1A1 and HO-1 also affected function, where the presence of CYP1A1 inhibited HO-1-mediated bilirubin formation by increasing the KmPOR HO-1 without affecting the Vmaxapp. Bilirubin 118-127 heme oxygenase 1 Homo sapiens 36-40 33394027-9 2021 This interaction between CYP1A1 and HO-1 also affected function, where the presence of CYP1A1 inhibited HO-1-mediated bilirubin formation by increasing the KmPOR HO-1 without affecting the Vmaxapp. Bilirubin 118-127 heme oxygenase 1 Homo sapiens 104-108 33394027-9 2021 This interaction between CYP1A1 and HO-1 also affected function, where the presence of CYP1A1 inhibited HO-1-mediated bilirubin formation by increasing the KmPOR HO-1 without affecting the Vmaxapp. Bilirubin 118-127 heme oxygenase 1 Homo sapiens 104-108 33394027-10 2021 In like manner, HO-1 inhibited CYP1A1-mediated 7-ethoxyresorufin dealkylation by increasing the KmPOR CYP1A1. ethoxyresorufin 47-64 heme oxygenase 1 Homo sapiens 16-20 33747471-0 2021 Intracellular ethanol-mediated oxidation and apoptosis in HepG2/CYP2E1 cells impaired by two active peptides from seahorse (Hippocampus kuda bleeler) protein hydrolysates via the Nrf2/HO-1 and akt pathways. Ethanol 14-21 heme oxygenase 1 Homo sapiens 184-188 33246336-15 2021 MAIN RESULTS AND THE ROLE OF CHANCE: After 1-week grafting, the relative expression of Sod1, Hmox1 and Cat was significantly higher in the group receiving 150 mg/kg NAC (NAC150-treated group) compared to controls (P = 0.04, P = 0.03, and P = 0.01, respectively), whereas the expression levels of Tnf-alpha, Il1-beta and Il6 were reduced. nac150 170-176 heme oxygenase 1 Homo sapiens 93-98 33502605-9 2021 Mechanically, aloin further enforced the activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling. alloin 14-19 heme oxygenase 1 Homo sapiens 89-105 33502605-9 2021 Mechanically, aloin further enforced the activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling. alloin 14-19 heme oxygenase 1 Homo sapiens 107-111 33585534-12 2020 LPP treatment improved cell survival rate, reduced intracellular LDH and MDA levels, increased intracellular SOD, CAT, GSH levels, down-regulated Bax, caspases-3, Nrf2, HO-1 expression, and up-regulated Bcl-2 expression. Linalyl diphosphate 0-3 heme oxygenase 1 Homo sapiens 169-173 33584308-0 2020 Mitochondrial Iron Overload-Mediated Inhibition of Nrf2-HO-1/GPX4 Assisted ALI-Induced Nephrotoxicity. Iron 14-18 heme oxygenase 1 Homo sapiens 56-60 33584308-0 2020 Mitochondrial Iron Overload-Mediated Inhibition of Nrf2-HO-1/GPX4 Assisted ALI-Induced Nephrotoxicity. aristolactam I 75-78 heme oxygenase 1 Homo sapiens 56-60 33584308-11 2020 In conclusion, our results demonstrated that mitochondrial iron overload-mediated antioxidant system inhibition would assist ALI-induced ferroptosis in renal tubular epithelial cells, and Nrf2-HO-1/GPX4 antioxidative system could be an important intervention target to prevent medicine containing ALI-induced nephropathy. aristolactam I 297-300 heme oxygenase 1 Homo sapiens 193-197 33472699-7 2021 In addition, TPP-Niacin markedly enhanced the expression of transcription factors (PGC-1alpha and NRF2) and antioxidant-associated genes (especially HO-1 and NQO-1). tpp-niacin 13-23 heme oxygenase 1 Homo sapiens 149-153 33472699-9 2021 TPP-Niacin is believed to protect against mitochondrial dysfunction by upregulating antioxidant-related genes, such as PGC-1alpha, NRF2, HO-1, and NQO-1, in RPE cells. tpp-niacin 0-10 heme oxygenase 1 Homo sapiens 137-141 33585534-0 2020 Protective Effect of Lemon Peel Polyphenols on Oxidative Stress-Induced Damage to Human Keratinocyte HaCaT Cells Through Activation of the Nrf2/HO-1 Signaling Pathway. Polyphenols 32-43 heme oxygenase 1 Homo sapiens 144-148 33505436-8 2020 Moreover, Zn-protoporphyrin-IX, an inhibitor of heme oxygenase-1, reduced the protective effect of Nrf1 overexpression on OGD/R-induced TEC oxidative stress and pyroptosis. zn-protoporphyrin-ix 10-30 heme oxygenase 1 Homo sapiens 48-64 33096358-10 2021 The present study further enriches and perfects the mechanism theory of HgCl2 toxicity and suggest that the CYP450 signaling and Nrf2/HO-1 pathway is important in shedding light on curcumin"s hepatoprotective effects in HgCl2 toxicity. Curcumin 181-189 heme oxygenase 1 Homo sapiens 134-138 33254827-8 2021 Simultaneously, flow cytometry also showed that NaHS could prevent mic-PS-induced accumulation of reactive oxygen species (ROS) by increasing the expression of HO-1 and NQO1. sodium bisulfide 48-52 heme oxygenase 1 Homo sapiens 160-164 33254827-8 2021 Simultaneously, flow cytometry also showed that NaHS could prevent mic-PS-induced accumulation of reactive oxygen species (ROS) by increasing the expression of HO-1 and NQO1. Reactive Oxygen Species 123-126 heme oxygenase 1 Homo sapiens 160-164 33254827-9 2021 Furthermore, inhibition of HO-1 could reverse the hepatic protective effects of NaHS during mic-PS exposure. sodium bisulfide 80-84 heme oxygenase 1 Homo sapiens 27-31 33254827-10 2021 Mechanistically, H2S elevating the HO-1 and NQO1 expression by facilitating nuclear accumulation of Nrf2, and consequently reducing mic-PS-induced hepatic apoptosis and inflammation. Deuterium 17-20 heme oxygenase 1 Homo sapiens 35-39 33278384-5 2021 This study was mainly aimed to explore whether the rhythm gene "brain and muscle ARNT-like 1" (BMAL1) was involved in regulating oxidative stress-related NRF2/HO-1 pathway to reduce the formation of urinary calcium oxalate stones. Calcium 207-214 heme oxygenase 1 Homo sapiens 159-163 33278384-6 2021 In vitro experiment found that the activation of NRF2/HO-1 can significantly reduce the oxalate-induced oxidative damage and urinary calcium oxalate stone formation, and the relative expression of BMAL1 was increased. Oxalates 88-95 heme oxygenase 1 Homo sapiens 54-58 33278384-6 2021 In vitro experiment found that the activation of NRF2/HO-1 can significantly reduce the oxalate-induced oxidative damage and urinary calcium oxalate stone formation, and the relative expression of BMAL1 was increased. Calcium 133-140 heme oxygenase 1 Homo sapiens 54-58 33278384-8 2021 In the hyperoxaluria animal model, the BMAL1 expression level decreased obviously, and the production of calcium oxalate stones was significantly reduced after activating NRF2/HO-1. Calcium Oxalate 105-120 heme oxygenase 1 Homo sapiens 176-180 33152749-3 2021 Here, we found that cell-free heme suppresses human B cell plasmablast/plasma cell differentiation by inhibiting the DOCK8/STAT3 signaling pathway, which is critical for B cell activation, as well as by upregulating heme oxygenase 1 (HO-1) through its enzymatic byproducts, carbon monoxide and biliverdin. Heme 30-34 heme oxygenase 1 Homo sapiens 216-232 33152749-3 2021 Here, we found that cell-free heme suppresses human B cell plasmablast/plasma cell differentiation by inhibiting the DOCK8/STAT3 signaling pathway, which is critical for B cell activation, as well as by upregulating heme oxygenase 1 (HO-1) through its enzymatic byproducts, carbon monoxide and biliverdin. Heme 30-34 heme oxygenase 1 Homo sapiens 234-238 33444090-7 2021 Under ICH pathological conditions, such as overproduction of reactive oxygen species (ROS), Nrf2 is translocated into the nucleus where it up-regulates the expression of several anti-oxidative phase II enzymes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 61-84 heme oxygenase 1 Homo sapiens 218-234 33444090-7 2021 Under ICH pathological conditions, such as overproduction of reactive oxygen species (ROS), Nrf2 is translocated into the nucleus where it up-regulates the expression of several anti-oxidative phase II enzymes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 61-84 heme oxygenase 1 Homo sapiens 236-240 33444090-7 2021 Under ICH pathological conditions, such as overproduction of reactive oxygen species (ROS), Nrf2 is translocated into the nucleus where it up-regulates the expression of several anti-oxidative phase II enzymes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 86-89 heme oxygenase 1 Homo sapiens 218-234 33444090-7 2021 Under ICH pathological conditions, such as overproduction of reactive oxygen species (ROS), Nrf2 is translocated into the nucleus where it up-regulates the expression of several anti-oxidative phase II enzymes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 86-89 heme oxygenase 1 Homo sapiens 236-240 33520087-7 2021 More importantly, berbamine increased the intracellular reactive oxygen species (ROS) level through the downregulation of antioxidative genes such as Nrf2, HO-1, SOD2, and GPX-1. berbamine 18-27 heme oxygenase 1 Homo sapiens 156-160 33520087-7 2021 More importantly, berbamine increased the intracellular reactive oxygen species (ROS) level through the downregulation of antioxidative genes such as Nrf2, HO-1, SOD2, and GPX-1. Reactive Oxygen Species 81-84 heme oxygenase 1 Homo sapiens 156-160 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Heme 97-101 heme oxygenase 1 Homo sapiens 0-16 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Heme 97-101 heme oxygenase 1 Homo sapiens 18-22 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Carbon Monoxide 166-181 heme oxygenase 1 Homo sapiens 0-16 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Carbon Monoxide 166-181 heme oxygenase 1 Homo sapiens 18-22 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Carbon Monoxide 183-185 heme oxygenase 1 Homo sapiens 0-16 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Carbon Monoxide 183-185 heme oxygenase 1 Homo sapiens 18-22 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Iron 193-197 heme oxygenase 1 Homo sapiens 0-16 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Iron 193-197 heme oxygenase 1 Homo sapiens 18-22 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Biliverdine 202-212 heme oxygenase 1 Homo sapiens 0-16 33440611-1 2021 Heme Oxygenase-1 (HO-1) is a type II detoxifying enzyme that catalyzes the rate-limiting step in heme degradation leading to the formation of equimolar quantities of carbon monoxide (CO), free iron and biliverdin. Biliverdine 202-212 heme oxygenase 1 Homo sapiens 18-22 33440611-6 2021 We provide a brief background on the current understanding of the mechanisms underlying how HO-1 leaves the sER membrane and migrates to the nucleus, the circumstances under which it does so and, maybe the most important and unknown aspect, what the function of HO-1 in the nucleus is. Serine 108-111 heme oxygenase 1 Homo sapiens 92-96 33271149-5 2021 Kaempferol (25 muM) mediated downregulation of GST, NQO1 and HO1 expression is also observed even after stimulation of Nrf2 by tert-butylhydroquinone (tBHQ). kaempferol 0-10 heme oxygenase 1 Homo sapiens 61-64 33096358-0 2021 Curcumin ameliorates mercuric chloride-induced liver injury via modulating cytochrome P450 signaling and Nrf2/HO-1 pathway. Curcumin 0-8 heme oxygenase 1 Homo sapiens 110-114 33096358-0 2021 Curcumin ameliorates mercuric chloride-induced liver injury via modulating cytochrome P450 signaling and Nrf2/HO-1 pathway. Mercuric Chloride 21-38 heme oxygenase 1 Homo sapiens 110-114 33096358-7 2021 Furthermore, curcumin significantly increased the nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and consequently upregulated the expression of heme oxygenase 1 (HO-1) under HgCl2 treatment. Curcumin 13-21 heme oxygenase 1 Homo sapiens 164-180 33096358-7 2021 Furthermore, curcumin significantly increased the nuclear translocation of nuclear factor E2-related factor 2 (Nrf2) and consequently upregulated the expression of heme oxygenase 1 (HO-1) under HgCl2 treatment. Curcumin 13-21 heme oxygenase 1 Homo sapiens 182-186 33096358-8 2021 Meanwhile, inhibition of HO-1 by zinc protoporphyria could abolish the cytoprotective effects of curcumin in HgCl2-treated L02 hepatocytes. Curcumin 97-105 heme oxygenase 1 Homo sapiens 25-29 33096358-8 2021 Meanwhile, inhibition of HO-1 by zinc protoporphyria could abolish the cytoprotective effects of curcumin in HgCl2-treated L02 hepatocytes. Mercuric Chloride 109-114 heme oxygenase 1 Homo sapiens 25-29 33096358-9 2021 In conclusion, our data identify that curcumin could enhance Nrf2-mediated HO-1 to upregulate antioxidant ability, which might be associate with CYP450 signaling to suppress liver damage induced by HgCl2. Curcumin 38-46 heme oxygenase 1 Homo sapiens 75-79 33628184-7 2020 Furthermore, Pristimerin suppressed NF-kappaB and MAPK signaling pathways, reduced reactive oxygen species (ROS) production and activated the nuclear factor erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signaling during osteoclastogenesis. pristimerin 13-24 heme oxygenase 1 Homo sapiens 209-213 33411783-11 2021 However, pretreatment of a HO-1 inhibitor reduced the protective effect of Formononetin on hyperoxic ALI. formononetin 75-87 heme oxygenase 1 Homo sapiens 27-31 33435366-8 2021 Chrysoeriol also markedly enhanced key transcription factors (Nrf2) and antioxidant-associated genes (particularly HO-1 and NQO-1). chrysoeriol 0-11 heme oxygenase 1 Homo sapiens 115-119 33411783-12 2021 Cell study showed that the Formononetin-induced upregulation of HO-1 was abolished when the Nrf2 was silenced. formononetin 27-39 heme oxygenase 1 Homo sapiens 64-68 33396673-0 2020 Dimethyl Fumarate Promotes the Survival of Retinal Ganglion Cells after Optic Nerve Injury, Possibly through the Nrf2/HO-1 Pathway. Dimethyl Fumarate 0-17 heme oxygenase 1 Homo sapiens 118-122 33411783-13 2021 CONCLUSIONS: Formononetin pretreatment reduces hyperoxia-induced ALI via Nrf2/HO-1-mediated antioxidant and anti-inflammatory effects. formononetin 13-25 heme oxygenase 1 Homo sapiens 78-82 33605427-11 2021 CONCLUSIONS: These results suggest that genistein exerts a neuroprotective effect on SH-SY5Y cells in vitro via Nrf2/HO-1/PI3K signaling, providing a foundation for the application of genistein in the treatment of neurodegenerative diseases related to Nrf2/HO-1/PI3K signaling. Genistein 40-49 heme oxygenase 1 Homo sapiens 117-121 33605427-11 2021 CONCLUSIONS: These results suggest that genistein exerts a neuroprotective effect on SH-SY5Y cells in vitro via Nrf2/HO-1/PI3K signaling, providing a foundation for the application of genistein in the treatment of neurodegenerative diseases related to Nrf2/HO-1/PI3K signaling. Genistein 40-49 heme oxygenase 1 Homo sapiens 257-261 33605427-11 2021 CONCLUSIONS: These results suggest that genistein exerts a neuroprotective effect on SH-SY5Y cells in vitro via Nrf2/HO-1/PI3K signaling, providing a foundation for the application of genistein in the treatment of neurodegenerative diseases related to Nrf2/HO-1/PI3K signaling. Genistein 184-193 heme oxygenase 1 Homo sapiens 117-121 33442403-0 2021 Inhibition of Nrf2/HO-1 signaling pathway by Dextran Sulfate suppresses angiogenesis of Gastric Cancer. Dextran Sulfate 45-60 heme oxygenase 1 Homo sapiens 19-23 32820550-6 2021 Other studies also demonstrated that the analgesic effects of opioids and cannabinoids are improved when these drugs are coadministered with CO-RMs, HO-1 or Nrf2 activators in different preclinical pain models and that these improvements are generally mediated by upregulation or prevention of the downregulation of micro-opioid receptors, delta-opioid receptors and/or cannabinoid 2 receptors in the setting of chronic pain. Cannabinoids 74-86 heme oxygenase 1 Homo sapiens 149-153 32820550-8 2021 In summary, activation of the Nrf2/HO-1/carbon monoxide signaling pathway alone and/or in combination with the administration of specific analgesics is a valid strategy for the treatment of chronic pain and some associated emotional disorders. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 35-39 33075721-0 2021 Role of heme oxygenase-1 in human placenta on iron supply to fetus. Iron 46-50 heme oxygenase 1 Homo sapiens 8-24 33075721-2 2021 Recently, increasing evidence has shown that heme oxygenase (HO)-1, which is an inducible isoform of the rate-limiting enzyme in the heme degradation pathway, may be involved in the effective reutilization of iron. Iron 209-213 heme oxygenase 1 Homo sapiens 45-66 33075721-9 2021 Placenta with fetal death (miscarriage) in the first and second trimester indicate significantly higher ratio of ho-1 gene for iron production to the fpn-1 gene for iron excretion than normal. Iron 127-131 heme oxygenase 1 Homo sapiens 113-117 33075721-11 2021 DISCUSSION: These findings suggest that HO-1 in placenta plays an important role in iron supplying system in the second trimester to support fetal development. Iron 84-88 heme oxygenase 1 Homo sapiens 40-44 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Heme 87-91 heme oxygenase 1 Homo sapiens 0-16 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Heme 87-91 heme oxygenase 1 Homo sapiens 18-22 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Heme 119-123 heme oxygenase 1 Homo sapiens 0-16 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Heme 119-123 heme oxygenase 1 Homo sapiens 18-22 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Carbon Monoxide 199-214 heme oxygenase 1 Homo sapiens 0-16 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Carbon Monoxide 199-214 heme oxygenase 1 Homo sapiens 18-22 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Biliverdine 219-229 heme oxygenase 1 Homo sapiens 0-16 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Biliverdine 219-229 heme oxygenase 1 Homo sapiens 18-22 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Bilirubin 230-239 heme oxygenase 1 Homo sapiens 0-16 33184238-4 2020 Heme oxygenase-1 (HO-1) is a cytoprotective, inducible enzyme that degrades toxic free heme released from destabilized heme proteins and, during this process, releases beneficial by-products such as carbon monoxide and biliverdin/bilirubin and promotes ferritin synthesis. Bilirubin 230-239 heme oxygenase 1 Homo sapiens 18-22 33383653-8 2020 Furthermore, cycloheximide treatment increased HO-1 abundance in p53 KO cells suggesting that p53 modulates HO-1 protein stability. Cycloheximide 13-26 heme oxygenase 1 Homo sapiens 47-51 33383653-8 2020 Furthermore, cycloheximide treatment increased HO-1 abundance in p53 KO cells suggesting that p53 modulates HO-1 protein stability. Cycloheximide 13-26 heme oxygenase 1 Homo sapiens 108-112 33289541-12 2020 TFNA treatment in IL-1beta-induced chondrocytes reduced apoptosis by activating the BCL2/BAX/caspase-3 pathway, inhibited oxidative stress by regulating the Nrf2/HO-1-signaling pathway, and enhanced autophagy through upregulated LC3-II, Beclin1, and Atg7. 4-(Trifluoromethyl)nicotinic acid 0-4 heme oxygenase 1 Homo sapiens 162-166 33414780-6 2020 Plasma levels of heme and its scavenger hemopexin and degrading enzyme heme-oxygenase-1 (HO-1) were measured in 48 STEC-HUS patients. Heme 17-21 heme oxygenase 1 Homo sapiens 89-93 33414780-10 2020 Interestingly, especially patients with high heme levels (n = 12, heme levels above 75 quartile range) had high plasma HO-1 levels with median of 332.5 (86-720) ng/ml (p = 0.008). Heme 45-49 heme oxygenase 1 Homo sapiens 119-123 33414780-10 2020 Interestingly, especially patients with high heme levels (n = 12, heme levels above 75 quartile range) had high plasma HO-1 levels with median of 332.5 (86-720) ng/ml (p = 0.008). Heme 66-70 heme oxygenase 1 Homo sapiens 119-123 33248029-10 2021 Modulation of oxidative stress-induced response involves the regulation of genes that encode for antioxidant proteins such as catalase and heme oxygenase-1; regulation that functions as a critical protection mechanism against oxidative cell damage induced by HEMA and TEGDMA. hydroxyethyl methacrylate 259-263 heme oxygenase 1 Homo sapiens 139-155 33248029-10 2021 Modulation of oxidative stress-induced response involves the regulation of genes that encode for antioxidant proteins such as catalase and heme oxygenase-1; regulation that functions as a critical protection mechanism against oxidative cell damage induced by HEMA and TEGDMA. triethylene glycol dimethacrylate 268-274 heme oxygenase 1 Homo sapiens 139-155 33220236-5 2021 Our data revealed that esculetin played an antioxidant role not only through its antioxidant activity, but also by highly inducing Nrf-2 translocation to the nucleus, which in turn, enhanced Nrf2 signaling regulated antioxidant genes (HO-1, NQO1, GCLM, SOD1 and SOD2) mRNA expression levels in H2O2-treated HCE cells. esculetin 23-32 heme oxygenase 1 Homo sapiens 235-239 33605427-0 2021 Genistein exerts a cell-protective effect via Nrf2/HO-1/PI3K signaling in Abeta25-35-induced Alzheimer"s disease models in vitro. Genistein 0-9 heme oxygenase 1 Homo sapiens 51-55 33605427-8 2021 RESULTS: Genistein enhanced the HO-1expression at both the mRNA and protein levels, as well as the PI3K p85 phosphorylation level. Genistein 9-18 heme oxygenase 1 Homo sapiens 32-36 33605427-9 2021 In addition, genistein increased the survival of SH-SY5Y cells treated with Abeta25-35via HO-1 signaling. Genistein 13-22 heme oxygenase 1 Homo sapiens 90-94 33442403-1 2021 Purpose: To investigate the role of Nrf2/HO-1 signaling pathway in angiogenesis and whether dextran sulfate (DS) could suppress angiogenesis by inhibiting Nrf2/HO-1 signaling pathway in gastric cancer. Dextran Sulfate 92-107 heme oxygenase 1 Homo sapiens 160-164 33442403-1 2021 Purpose: To investigate the role of Nrf2/HO-1 signaling pathway in angiogenesis and whether dextran sulfate (DS) could suppress angiogenesis by inhibiting Nrf2/HO-1 signaling pathway in gastric cancer. Dextran Sulfate 109-111 heme oxygenase 1 Homo sapiens 160-164 33442403-4 2021 Immunofluorescence,western blot and qPCR analyzed the effects of DS on the expression of Nrf2, HO-1 and VEGF under gradient hypoxia time. Dextran Sulfate 65-67 heme oxygenase 1 Homo sapiens 95-99 33442403-10 2021 DS reduced the angiogenic potential and the expression of Nrf2, HO-1 and VEGF in HGC-27 cells. Dextran Sulfate 0-2 heme oxygenase 1 Homo sapiens 64-68 33357768-0 2021 MitoQ protects against high glucose-induced brain microvascular endothelial cells injury via the Nrf2/HO-1 pathway. mitoquinone 0-5 heme oxygenase 1 Homo sapiens 102-106 33357768-6 2021 Moreover, we found that MitoQ treatment significantly upregulated the expression of Nrf2 and HO-1 in HG-induced BMECs, which is accompanied by improved mitochondrial membrane potential and decreased MtROS production. mitoquinone 24-29 heme oxygenase 1 Homo sapiens 93-97 33357768-9 2021 In conclusion, our results suggest that MitoQ exerts protective effect on HG-induced BMECs injury via activating Nrf2/HO-1 pathway. mitoquinone 40-45 heme oxygenase 1 Homo sapiens 118-122 33357768-9 2021 In conclusion, our results suggest that MitoQ exerts protective effect on HG-induced BMECs injury via activating Nrf2/HO-1 pathway. Mercury 74-76 heme oxygenase 1 Homo sapiens 118-122 33373320-5 2020 NMN administration significantly increased NAD+ levels, SIRT1 protein expression, and heme oxygenase-1 (HO-1) expression. Nicotinamide Mononucleotide 0-3 heme oxygenase 1 Homo sapiens 86-102 33373320-5 2020 NMN administration significantly increased NAD+ levels, SIRT1 protein expression, and heme oxygenase-1 (HO-1) expression. Nicotinamide Mononucleotide 0-3 heme oxygenase 1 Homo sapiens 104-108 33396673-1 2020 This study aimed to verify whether dimethyl fumarate (DMF) promotes the survival of retinal ganglion cells (RGCs) after optic nerve crush (ONC) accompanied by activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Dimethyl Fumarate 35-52 heme oxygenase 1 Homo sapiens 207-223 33396673-1 2020 This study aimed to verify whether dimethyl fumarate (DMF) promotes the survival of retinal ganglion cells (RGCs) after optic nerve crush (ONC) accompanied by activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Dimethyl Fumarate 35-52 heme oxygenase 1 Homo sapiens 225-229 33396673-1 2020 This study aimed to verify whether dimethyl fumarate (DMF) promotes the survival of retinal ganglion cells (RGCs) after optic nerve crush (ONC) accompanied by activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Dimethyl Fumarate 54-57 heme oxygenase 1 Homo sapiens 207-223 33396673-1 2020 This study aimed to verify whether dimethyl fumarate (DMF) promotes the survival of retinal ganglion cells (RGCs) after optic nerve crush (ONC) accompanied by activation of the NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway. Dimethyl Fumarate 54-57 heme oxygenase 1 Homo sapiens 225-229 33396673-3 2020 Furthermore, immunohistochemical and immunoblotting analyses were performed to study the activation of the Nrf2/HO-1 pathway using retinas treated with daily administration of DMF. Dimethyl Fumarate 176-179 heme oxygenase 1 Homo sapiens 112-116 33396673-5 2020 Immunohistochemical analysis showed that DMF administration increased the immunoreactivity for Nrf2 and HO-1, a potent antioxidant enzyme, in RGCs immunolabeled with RNA-binding protein with multiple splicing (RBPMS). Dimethyl Fumarate 41-44 heme oxygenase 1 Homo sapiens 104-108 33396673-6 2020 Immunoblotting analysis revealed an increase in the nuclear expression of Nrf2 and marked upregulation of HO-1 after DMF administration. Dimethyl Fumarate 117-120 heme oxygenase 1 Homo sapiens 106-110 33344146-9 2020 In addition, intracellular cholesterol load interfered with the balance between NADPH oxidase activity and HO-1 expression. Cholesterol 27-38 heme oxygenase 1 Homo sapiens 107-111 33051205-1 2020 Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Heme 45-49 heme oxygenase 1 Homo sapiens 31-34 33051205-1 2020 Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Biliverdine 65-75 heme oxygenase 1 Homo sapiens 31-34 33051205-1 2020 Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Carbon Monoxide 77-79 heme oxygenase 1 Homo sapiens 31-34 33051205-1 2020 Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Iron 85-89 heme oxygenase 1 Homo sapiens 31-34 33051205-1 2020 Heme oxygenase-2 (HO2) and -1 (HO1) catalyze heme degradation to biliverdin, CO, and iron, forming an essential link in the heme metabolism network. Heme 124-128 heme oxygenase 1 Homo sapiens 31-34 33051205-2 2020 Tight regulation of the cellular levels and catalytic activities of HO1 and HO2 is important for maintaining heme homeostasis. Heme 109-113 heme oxygenase 1 Homo sapiens 68-71 33287198-4 2020 According to the results, PGFE suppressed pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukin (IL)-1beta, and IL-6, and pro-inflammatory mediators such as inducible nitric oxide synthase and cyclooxygenase-2 through heme oxygenase-1 expression in human periodontal ligament cells stimulated with Porphyromonas gingivalis lipopolysaccharide (PG-LPS). pgfe 26-30 heme oxygenase 1 Homo sapiens 240-256 33333908-0 2020 Novel Heme Oxygenase-1 (HO-1) Inducers Based on Dimethyl Fumarate Structure. Dimethyl Fumarate 48-65 heme oxygenase 1 Homo sapiens 6-22 33333908-0 2020 Novel Heme Oxygenase-1 (HO-1) Inducers Based on Dimethyl Fumarate Structure. Dimethyl Fumarate 48-65 heme oxygenase 1 Homo sapiens 24-28 33333908-1 2020 Novel heme oxygenase-1 (HO-1) inducers based on dimethyl fumarate (DMF) structure are reported in this paper. Dimethyl Fumarate 48-65 heme oxygenase 1 Homo sapiens 6-22 33333908-1 2020 Novel heme oxygenase-1 (HO-1) inducers based on dimethyl fumarate (DMF) structure are reported in this paper. Dimethyl Fumarate 48-65 heme oxygenase 1 Homo sapiens 24-28 33333908-1 2020 Novel heme oxygenase-1 (HO-1) inducers based on dimethyl fumarate (DMF) structure are reported in this paper. Dimethyl Fumarate 67-70 heme oxygenase 1 Homo sapiens 6-22 33333908-1 2020 Novel heme oxygenase-1 (HO-1) inducers based on dimethyl fumarate (DMF) structure are reported in this paper. Dimethyl Fumarate 67-70 heme oxygenase 1 Homo sapiens 24-28 33327226-8 2020 The increase in heme in vivo induces HO-1 production, a heme-degrading enzyme. Heme 16-20 heme oxygenase 1 Homo sapiens 37-41 33327226-8 2020 The increase in heme in vivo induces HO-1 production, a heme-degrading enzyme. Heme 56-60 heme oxygenase 1 Homo sapiens 37-41 33343802-0 2020 Induction of HO-1 by 5, 8-Dihydroxy-4",7-Dimethoxyflavone via Activation of ROS/p38 MAPK/Nrf2 Attenuates Thrombin-Induced Connective Tissue Growth Factor Expression in Human Cardiac Fibroblasts. 5, 8-dihydroxy-4",7-dimethoxyflavone 21-57 heme oxygenase 1 Homo sapiens 13-17 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 0-16 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 18-22 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Carbon Monoxide 111-113 heme oxygenase 1 Homo sapiens 0-16 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Carbon Monoxide 111-113 heme oxygenase 1 Homo sapiens 18-22 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Iron 121-125 heme oxygenase 1 Homo sapiens 0-16 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Iron 121-125 heme oxygenase 1 Homo sapiens 18-22 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Biliverdine 131-141 heme oxygenase 1 Homo sapiens 0-16 33276470-1 2020 Heme oxygenase 1 (HO-1) is the rate-limiting enzyme of heme oxidative degradation, generating carbon monoxide (CO), free iron, and biliverdin. Biliverdine 131-141 heme oxygenase 1 Homo sapiens 18-22 31851841-13 2020 Furthermore, taraxasterol induced the Nrf2 nuclear accumulation and expressions of HO-1, NQO-1 and GPx-3 in OGD/R-induced hippocampal neurons. taraxasterol 13-25 heme oxygenase 1 Homo sapiens 83-87 32954935-9 2020 Recombinant DEL-1 augmented AMP-activated protein kinase (AMPK) phosphorylation and haem oxygenase (HO)-1 expression to alleviating inflammation and impairment of insulin signalling in 3T3-L1 adipocytes treated with palmitate. Palmitates 216-225 heme oxygenase 1 Homo sapiens 84-105 33343802-0 2020 Induction of HO-1 by 5, 8-Dihydroxy-4",7-Dimethoxyflavone via Activation of ROS/p38 MAPK/Nrf2 Attenuates Thrombin-Induced Connective Tissue Growth Factor Expression in Human Cardiac Fibroblasts. Reactive Oxygen Species 76-79 heme oxygenase 1 Homo sapiens 13-17 33343802-2 2020 Flavonoids have been demonstrated to display anti-inflammatory and antioxidant effects through the induction of HO-1. Flavonoids 0-10 heme oxygenase 1 Homo sapiens 112-116 33343802-5 2020 Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. Reactive Oxygen Species 144-167 heme oxygenase 1 Homo sapiens 68-72 33343802-5 2020 Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. Reactive Oxygen Species 169-172 heme oxygenase 1 Homo sapiens 68-72 33343802-5 2020 Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. Acetylcysteine 184-201 heme oxygenase 1 Homo sapiens 68-72 33343802-5 2020 Here, we found that DDF time- and concentration-dependently induced HO-1 protein and mRNA expression, which was attenuated by pretreatment with reactive oxygen species (ROS) scavenger N-acetyl cysteine (NAC) in HCFs. Acetylcysteine 203-206 heme oxygenase 1 Homo sapiens 68-72 33343802-7 2020 Interestingly, pretreatment with glutathione (GSH) inhibited DDF-induced HO-1 expression. Glutathione 33-44 heme oxygenase 1 Homo sapiens 73-77 33343802-7 2020 Interestingly, pretreatment with glutathione (GSH) inhibited DDF-induced HO-1 expression. Glutathione 46-49 heme oxygenase 1 Homo sapiens 73-77 33343802-7 2020 Interestingly, pretreatment with glutathione (GSH) inhibited DDF-induced HO-1 expression. ddf 61-64 heme oxygenase 1 Homo sapiens 73-77 33343802-9 2020 DDF-induced HO-1 expression was attenuated by an inhibitor of p38 MAPK (p38i VIII) or siRNA, but not by MEK1/2 (PD98059) or JNK1/2 (SP600125). ddf 0-3 heme oxygenase 1 Homo sapiens 12-16 33343802-11 2020 Moreover, DDF-induced HO-1 expression was mediated through Nrf2 phosphorylation and translocation into the nucleus which was attenuated by NAC or p38 siRNA. Acetylcysteine 139-142 heme oxygenase 1 Homo sapiens 22-26 33343802-13 2020 Interaction between Nrf2 and the ARE-binding sites on the HO-1 promoter was revealed by chromatin immunoprecipitation assay, which was attenuated by NAC, GSH, or p38i VIII. Acetylcysteine 149-152 heme oxygenase 1 Homo sapiens 58-62 33343802-13 2020 Interaction between Nrf2 and the ARE-binding sites on the HO-1 promoter was revealed by chromatin immunoprecipitation assay, which was attenuated by NAC, GSH, or p38i VIII. Glutathione 154-157 heme oxygenase 1 Homo sapiens 58-62 33343802-16 2020 These results suggested that DDF-induced HO-1 expression is, at least, mediated through the activation of the ROS-dependent p38 MAPK/Nrf2 signaling pathway in HCFs. Reactive Oxygen Species 110-113 heme oxygenase 1 Homo sapiens 41-45 32623920-6 2020 We further showed that H2O2-induced oxidative stress was reduced by curcumin via the Nrf2/HO-1 pathway in human neuroblastoma cells. Hydrogen Peroxide 23-27 heme oxygenase 1 Homo sapiens 90-94 33199994-9 2020 DMF also increased the anti-inflammatory and antioxidative ability of NP cells by promoting the activity of the Nrf2/HO-1 and PI3K/Akt signaling pathways, thus delaying IVDD. Dimethyl Fumarate 0-3 heme oxygenase 1 Homo sapiens 117-121 33378016-0 2020 MiRNA-15a-3p inhibits the metastasis of hepatocellular carcinoma by interacting with HMOX1. mirna-15a-3p 0-12 heme oxygenase 1 Homo sapiens 85-90 32623920-6 2020 We further showed that H2O2-induced oxidative stress was reduced by curcumin via the Nrf2/HO-1 pathway in human neuroblastoma cells. Curcumin 68-76 heme oxygenase 1 Homo sapiens 90-94 33045867-9 2020 Subsequently, activation of MAPK-Nrf2/ARE signaling was readily induced by vitexin treatment, as evidenced by upregulation of antioxidant genes including heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). vitexin 75-82 heme oxygenase 1 Homo sapiens 154-170 33045867-9 2020 Subsequently, activation of MAPK-Nrf2/ARE signaling was readily induced by vitexin treatment, as evidenced by upregulation of antioxidant genes including heme oxygenase 1 (HO-1) and superoxide dismutase (SOD). vitexin 75-82 heme oxygenase 1 Homo sapiens 172-176 33035580-0 2020 Effects of taraxasterol against ethanol and high-fat diet-induced liver injury by regulating TLR4/MyD88/NF-kappaB and Nrf2/HO-1 signaling pathways. taraxasterol 11-23 heme oxygenase 1 Homo sapiens 123-127 33456997-11 2020 Crocin also increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), decreased TAZ in MNNG-treated GES-1 cells. crocin 0-6 heme oxygenase 1 Homo sapiens 95-111 33456997-11 2020 Crocin also increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), decreased TAZ in MNNG-treated GES-1 cells. crocin 0-6 heme oxygenase 1 Homo sapiens 113-117 33035580-6 2020 Production of CYP2E1, Nrf2 and HO-1, which produced in response to increased oxidative stress, were regulated in TAR treated, ethanol-induced hepatocytes. Ethanol 126-133 heme oxygenase 1 Homo sapiens 31-35 33256538-0 2022 Aloperine protects human retinal pigment epithelial cells against hydrogen peroxide-induced oxidative stress and apoptosis through activation of Nrf2/HO-1 pathway. aloperine 0-9 heme oxygenase 1 Homo sapiens 150-154 32942007-5 2020 Moreover, we evaluated the relationship between glucose concentration and concentration of intracellular ROS, as well as the effects of glucose concentration on the induction of Nrf2-dependent genes such as Glutathione S-transferases, Heme-oxygenase-1, and Glutathione peroxidase-4). Glucose 136-143 heme oxygenase 1 Homo sapiens 235-251 33157356-5 2020 HO-1 induction by transfection of HO-1 over-expression plasmid or treatment with cobalt protoporphyrin (CoPP), a potent HO-1 inducer, could inhibit DTMUV replication effectively. cobaltiprotoporphyrin 81-102 heme oxygenase 1 Homo sapiens 0-4 33157356-5 2020 HO-1 induction by transfection of HO-1 over-expression plasmid or treatment with cobalt protoporphyrin (CoPP), a potent HO-1 inducer, could inhibit DTMUV replication effectively. cobaltiprotoporphyrin 104-108 heme oxygenase 1 Homo sapiens 0-4 33157356-7 2020 Furthermore, we found that ferric ion (Fe3+) but not biliverdin and carbon monoxide, products of heme degradation by HO-1, mediated the HO-1-induced anti-DTMUV effect. Ferric enterobactin ion 27-33 heme oxygenase 1 Homo sapiens 136-140 33157356-7 2020 Furthermore, we found that ferric ion (Fe3+) but not biliverdin and carbon monoxide, products of heme degradation by HO-1, mediated the HO-1-induced anti-DTMUV effect. ferric sulfate 39-43 heme oxygenase 1 Homo sapiens 136-140 33157356-7 2020 Furthermore, we found that ferric ion (Fe3+) but not biliverdin and carbon monoxide, products of heme degradation by HO-1, mediated the HO-1-induced anti-DTMUV effect. Heme 97-101 heme oxygenase 1 Homo sapiens 136-140 33256538-0 2022 Aloperine protects human retinal pigment epithelial cells against hydrogen peroxide-induced oxidative stress and apoptosis through activation of Nrf2/HO-1 pathway. Hydrogen Peroxide 66-83 heme oxygenase 1 Homo sapiens 150-154 33256538-15 2022 Moreover, aloperine treatment enhanced the expression levels of Nrf2 in nuclear fraction and the HO-1 expression in lysates. aloperine 10-19 heme oxygenase 1 Homo sapiens 97-101 33256538-17 2022 In conclusion, these findings demonstrated that aloperine protected ARPE-19 cells from H2O2-induced oxidative stress and apoptosis in part via activating the Nrf2/HO-1 signaling pathway. aloperine 48-57 heme oxygenase 1 Homo sapiens 163-167 33256538-17 2022 In conclusion, these findings demonstrated that aloperine protected ARPE-19 cells from H2O2-induced oxidative stress and apoptosis in part via activating the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 87-91 heme oxygenase 1 Homo sapiens 163-167 32812103-4 2020 Additionally, coumarin-pi promoted nuclear factor erythroid 2-related factor 2 (Nrf2) expression and upregulated the protein expression of antioxidantenzymes, including heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidase (NQO1), glutamyl cysteine ligase catalytic subunit (GCLC), and glutamate-cysteine ligase regulatory subunit (GCLM). coumarin 14-22 heme oxygenase 1 Homo sapiens 169-185 32812103-4 2020 Additionally, coumarin-pi promoted nuclear factor erythroid 2-related factor 2 (Nrf2) expression and upregulated the protein expression of antioxidantenzymes, including heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidase (NQO1), glutamyl cysteine ligase catalytic subunit (GCLC), and glutamate-cysteine ligase regulatory subunit (GCLM). coumarin 14-22 heme oxygenase 1 Homo sapiens 187-191 32976958-7 2020 Both HFD-fed mice and PA/OA-induced HepG2 cells displayed ferroptosis-based panel of biomarkers such as iron overload with the up-regulation of TFR1 and the down-regulation of FTH1, lipid peroxidation and inhibition of Nrf2 activity, which further induced GPX4 and HO-1 levels. Iron 104-108 heme oxygenase 1 Homo sapiens 265-269 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Biliverdine 99-109 heme oxygenase 1 Homo sapiens 0-16 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Biliverdine 99-109 heme oxygenase 1 Homo sapiens 18-22 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Iron 111-115 heme oxygenase 1 Homo sapiens 0-16 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Iron 111-115 heme oxygenase 1 Homo sapiens 18-22 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 0-16 33266044-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme molecules releasing equimolar amounts of biliverdin, iron and carbon monoxide. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 18-22 33381031-0 2020 Suppression of Urokinase-Type Plasminogen Activator Receptor by Docosahexaenoic Acid Mediated by Heme Oxygenase-1 in 12-O-Tetradecanoylphorbol-13-Acetate-Induced Human Endothelial Cells. Docosahexaenoic Acids 64-84 heme oxygenase 1 Homo sapiens 97-113 33260980-1 2020 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that catalyzes heme group degradation. Heme 74-78 heme oxygenase 1 Homo sapiens 0-16 33260980-1 2020 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that catalyzes heme group degradation. Heme 74-78 heme oxygenase 1 Homo sapiens 18-22 33381031-0 2020 Suppression of Urokinase-Type Plasminogen Activator Receptor by Docosahexaenoic Acid Mediated by Heme Oxygenase-1 in 12-O-Tetradecanoylphorbol-13-Acetate-Induced Human Endothelial Cells. Tetradecanoylphorbol Acetate 117-153 heme oxygenase 1 Homo sapiens 97-113 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). fepp 47-51 heme oxygenase 1 Homo sapiens 13-17 33381031-4 2020 The aim of this study was to investigate the effect of DHA on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced uPAR expression and potential role of heme oxygenase-1 (HO-1) in DHA-induced inhibition of uPAR in human endothelial ECV304 cells. Docosahexaenoic Acids 178-181 heme oxygenase 1 Homo sapiens 151-167 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). fepp 47-51 heme oxygenase 1 Homo sapiens 144-148 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). Tetradecanoylphorbol Acetate 63-66 heme oxygenase 1 Homo sapiens 13-17 33381031-4 2020 The aim of this study was to investigate the effect of DHA on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced uPAR expression and potential role of heme oxygenase-1 (HO-1) in DHA-induced inhibition of uPAR in human endothelial ECV304 cells. Docosahexaenoic Acids 178-181 heme oxygenase 1 Homo sapiens 169-173 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). Tetradecanoylphorbol Acetate 63-66 heme oxygenase 1 Homo sapiens 144-148 33381031-6 2020 Moreover, treatment with DHA induced HO-1 expression in a time- and concentration-dependent manner. Docosahexaenoic Acids 25-28 heme oxygenase 1 Homo sapiens 37-41 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). tin protoporphyrin IX 159-180 heme oxygenase 1 Homo sapiens 13-17 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). tin protoporphyrin IX 159-180 heme oxygenase 1 Homo sapiens 144-148 33381031-7 2020 In addition, DHA-induced inhibition of uPAR expression and cell invasion in TPA-stimulated cells was reversed by si-HO-1 RNA. Docosahexaenoic Acids 13-16 heme oxygenase 1 Homo sapiens 116-120 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 182-186 heme oxygenase 1 Homo sapiens 13-17 33381031-7 2020 In addition, DHA-induced inhibition of uPAR expression and cell invasion in TPA-stimulated cells was reversed by si-HO-1 RNA. Tetradecanoylphorbol Acetate 76-79 heme oxygenase 1 Homo sapiens 116-120 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 182-186 heme oxygenase 1 Homo sapiens 144-148 33381031-8 2020 Induction of HO-1 by ferric protoporphyrin IX (FePP) inhibited TPA-induced uPAR expression, and this effect was abolished by treatment with the HO-1 inhibitor tin protoporphyrin IX (SnPP). ferric protoporphyrin ix 21-45 heme oxygenase 1 Homo sapiens 13-17 33381031-9 2020 Additionally, carbon monoxide, an HO-1 product, attenuated TPA-induced uPAR expression and cell invasion. Carbon Monoxide 14-29 heme oxygenase 1 Homo sapiens 34-38 33381031-9 2020 Additionally, carbon monoxide, an HO-1 product, attenuated TPA-induced uPAR expression and cell invasion. Tetradecanoylphorbol Acetate 59-62 heme oxygenase 1 Homo sapiens 34-38 33381031-10 2020 Collectively, these data suggest a novel role of DHA-induced HO-1 in reducing uPAR expression and cell invasion in human endothelial ECV304 cells. Docosahexaenoic Acids 49-52 heme oxygenase 1 Homo sapiens 61-65 33238962-7 2020 Serum HO-1 correlated with serum total bilirubin (R = 0.454, P < 0.001) and serum LDH (R = 0.500, P < 0.001). Bilirubin 39-48 heme oxygenase 1 Homo sapiens 6-10 33232319-0 2020 LncRNA MIR4435-2HG mediates cisplatin resistance in HCT116 cells by regulating Nrf2 and HO-1. Cisplatin 28-37 heme oxygenase 1 Homo sapiens 88-92 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. mir4435 130-137 heme oxygenase 1 Homo sapiens 22-26 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. mir4435 130-137 heme oxygenase 1 Homo sapiens 225-229 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. mir4435 159-166 heme oxygenase 1 Homo sapiens 22-26 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. mir4435 159-166 heme oxygenase 1 Homo sapiens 225-229 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. Cisplatin 180-189 heme oxygenase 1 Homo sapiens 22-26 33232319-9 2020 Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. Cisplatin 180-189 heme oxygenase 1 Homo sapiens 225-229 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Heme 98-102 heme oxygenase 1 Homo sapiens 0-16 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. zinc protoporphyrin 56-78 heme oxygenase 1 Homo sapiens 22-38 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. zinc protoporphyrin 56-78 heme oxygenase 1 Homo sapiens 40-44 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. zinc protoporphyrin 80-87 heme oxygenase 1 Homo sapiens 22-38 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. zinc protoporphyrin 80-87 heme oxygenase 1 Homo sapiens 40-44 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. Edaravone 147-156 heme oxygenase 1 Homo sapiens 22-38 33222098-8 2022 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 muM) smothered the preventive impacts brought about by edaravone with respect to mitochondrial function and inflammation. Edaravone 147-156 heme oxygenase 1 Homo sapiens 40-44 33222098-9 2022 After this examination, it can be concluded that edaravone caused cytoprotective impacts in an HO-1-dependent manner in SH-SY5Y cells against ZnO NPs-induced toxicity. Edaravone 49-58 heme oxygenase 1 Homo sapiens 95-99 33222098-9 2022 After this examination, it can be concluded that edaravone caused cytoprotective impacts in an HO-1-dependent manner in SH-SY5Y cells against ZnO NPs-induced toxicity. Zinc Oxide 142-145 heme oxygenase 1 Homo sapiens 95-99 33274001-7 2020 In conclusion, our study suggested that delphinidin, as an effective antioxidant, protected HepG2 cells from oxidative stress by regulating the expression of Nrf2/HO-1. delphinidin 40-51 heme oxygenase 1 Homo sapiens 163-167 33228202-8 2020 HO-1 degrades the prooxidant heme and generates molecules with antioxidative and anti-inflammatory properties, resulting in decreased oxidative stress and inflammation. Heme 29-33 heme oxygenase 1 Homo sapiens 0-4 32900489-6 2020 Furthermore, ADA modulated the expressions of SOD2, HO-1 and Gpx1 as antioxidant enzymes. adrenic acid 13-16 heme oxygenase 1 Homo sapiens 52-56 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Heme 98-102 heme oxygenase 1 Homo sapiens 18-22 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Carbon Monoxide 106-121 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Carbon Monoxide 106-121 heme oxygenase 1 Homo sapiens 18-22 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Carbon Monoxide 123-125 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Carbon Monoxide 123-125 heme oxygenase 1 Homo sapiens 18-22 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Iron 128-132 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Iron 128-132 heme oxygenase 1 Homo sapiens 18-22 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Biliverdine 138-148 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Biliverdine 138-148 heme oxygenase 1 Homo sapiens 18-22 33210267-2 2021 The current study, demonstrated that 5-ALA induced ferroptosis via glutathione peroxidase 4 (GPX4) and heme oxygenase 1 (HMOX1) and had an antitumor effect in esophageal squamous cell carcinoma (ESCC). 5-amino levulinic acid 37-42 heme oxygenase 1 Homo sapiens 103-119 33210267-2 2021 The current study, demonstrated that 5-ALA induced ferroptosis via glutathione peroxidase 4 (GPX4) and heme oxygenase 1 (HMOX1) and had an antitumor effect in esophageal squamous cell carcinoma (ESCC). 5-amino levulinic acid 37-42 heme oxygenase 1 Homo sapiens 121-126 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Biliverdine 150-152 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Biliverdine 150-152 heme oxygenase 1 Homo sapiens 18-22 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Bilirubin 191-200 heme oxygenase 1 Homo sapiens 0-16 33228260-1 2020 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the oxidative conversion of heme to carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is converted to bilirubin (BR) by biliverdin reductase. Bilirubin 191-200 heme oxygenase 1 Homo sapiens 18-22 33228260-4 2020 HO-1 may function as a pleiotropic modulator of inflammatory signaling, via the removal of heme, and generation of its enzymatic degradation-products. Heme 91-95 heme oxygenase 1 Homo sapiens 0-4 33185862-0 2021 Piperlongumine Inhibits Zika Virus Replication In vitro and Promotes Up-Regulation of HO-1 Expression, Suggesting An Implication of Oxidative Stress. piperlonguminine 0-14 heme oxygenase 1 Homo sapiens 86-90 33217990-6 2020 Curcumin quenches free radicals, induces antioxidant enzymes (catalase, superoxide dismutase, glutathione peroxidase), and upregulates antioxidative protein markers-Nrf2 and HO-1 that lead to the suppression of cellular oxidative stress. Curcumin 0-8 heme oxygenase 1 Homo sapiens 174-178 33202598-10 2020 Moreover, chronic obesity also results in the reduction of antioxidant genes such as heme oxygenase-1 (HO-1), increasing adipocyte susceptibility to ROS and cytokines. Reactive Oxygen Species 149-152 heme oxygenase 1 Homo sapiens 85-101 33223823-0 2020 Eldecalcitol Inhibits LPS-Induced NLRP3 Inflammasome-Dependent Pyroptosis in Human Gingival Fibroblasts by Activating the Nrf2/HO-1 Signaling Pathway. eldecalcitol 0-12 heme oxygenase 1 Homo sapiens 127-131 33223823-12 2020 Pretreatment with ED-71 effectively inhibited the LPS-induced pyroptosis and was associated with activation of the Nrf2/HO-1 signaling pathway. eldecalcitol 18-23 heme oxygenase 1 Homo sapiens 120-124 33294119-0 2020 Transcriptome Investigation and In Vitro Verification of Curcumin-Induced HO-1 as a Feature of Ferroptosis in Breast Cancer Cells. Curcumin 57-65 heme oxygenase 1 Homo sapiens 74-78 33294119-7 2020 Curcumin upregulates a variety of ferroptosis target genes related to redox regulation, especially heme oxygenase-1 (HO-1). Curcumin 0-8 heme oxygenase 1 Homo sapiens 99-115 33294119-7 2020 Curcumin upregulates a variety of ferroptosis target genes related to redox regulation, especially heme oxygenase-1 (HO-1). Curcumin 0-8 heme oxygenase 1 Homo sapiens 117-121 33294119-9 2020 Therefore, these data demonstrate that curcumin triggers the molecular and cytological characteristics of ferroptosis in breast cancer cells, and HO-1 promotes curcumin-induced ferroptosis. Curcumin 160-168 heme oxygenase 1 Homo sapiens 146-150 33185862-5 2021 PL also significantly increased heme oxygenase-1 (HO-1) gene expression, while silencing HO-1 expression and using the reactive oxygen species scavenger, N-acetylcysteine, attenuated the inhibitory effect of PL on Zika virus replication. piperlonguminine 0-2 heme oxygenase 1 Homo sapiens 32-48 33185862-5 2021 PL also significantly increased heme oxygenase-1 (HO-1) gene expression, while silencing HO-1 expression and using the reactive oxygen species scavenger, N-acetylcysteine, attenuated the inhibitory effect of PL on Zika virus replication. piperlonguminine 0-2 heme oxygenase 1 Homo sapiens 50-54 33185862-5 2021 PL also significantly increased heme oxygenase-1 (HO-1) gene expression, while silencing HO-1 expression and using the reactive oxygen species scavenger, N-acetylcysteine, attenuated the inhibitory effect of PL on Zika virus replication. piperlonguminine 208-210 heme oxygenase 1 Homo sapiens 89-93 32885420-0 2020 Protective role of HO-1 against acute kidney injury caused by cutaneous exposure to arsenicals. Arsenicals 84-94 heme oxygenase 1 Homo sapiens 19-23 33148696-11 2021 Whereas BRET data suggested substantial inhibition of CYP1A2-mediated 7-ethoxyresorufin deethylation in the presence of HO-1, its activity was actually stimulated at subsaturating POR. ethoxyresorufin 70-87 heme oxygenase 1 Homo sapiens 120-124 33148696-12 2021 In contrast, HO-1-mediated heme metabolism was inhibited at subsaturating POR. Heme 27-31 heme oxygenase 1 Homo sapiens 13-17 33959213-5 2020 In THP-1 macrophage-derived foam cells, treatment with BCA upregulated ATP-binding cassette (ABC) transporter A1 (ABCA1) and ABCG1 expression and facilitated subsequent cholesterol efflux and diminished intracellular cholesterol contents by activating the peroxisome proliferator-activated receptor gamma (PPARgamma)/liver X receptor alpha (LXRalpha) and PPARgamma/heme oxygenase 1 (HO-1) pathways. biochanin A 55-58 heme oxygenase 1 Homo sapiens 355-381 33959213-5 2020 In THP-1 macrophage-derived foam cells, treatment with BCA upregulated ATP-binding cassette (ABC) transporter A1 (ABCA1) and ABCG1 expression and facilitated subsequent cholesterol efflux and diminished intracellular cholesterol contents by activating the peroxisome proliferator-activated receptor gamma (PPARgamma)/liver X receptor alpha (LXRalpha) and PPARgamma/heme oxygenase 1 (HO-1) pathways. biochanin A 55-58 heme oxygenase 1 Homo sapiens 383-387 33174867-7 2020 Importantly, memantine also promoted the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) antioxidant signaling pathway. Memantine 13-22 heme oxygenase 1 Homo sapiens 110-126 33174867-7 2020 Importantly, memantine also promoted the expression of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) antioxidant signaling pathway. Memantine 13-22 heme oxygenase 1 Homo sapiens 128-132 32885420-8 2020 Indeed, HO-1 induction with cobalt protoporphyrin (CoPP) treatment in arsenical-treated HEK293 cells afforded cytoprotection by attenuating CHOP-associated apoptosis and cytokine mRNA levels. cobaltiprotoporphyrin 28-49 heme oxygenase 1 Homo sapiens 8-12 32885420-8 2020 Indeed, HO-1 induction with cobalt protoporphyrin (CoPP) treatment in arsenical-treated HEK293 cells afforded cytoprotection by attenuating CHOP-associated apoptosis and cytokine mRNA levels. cobaltiprotoporphyrin 51-55 heme oxygenase 1 Homo sapiens 8-12 32885420-8 2020 Indeed, HO-1 induction with cobalt protoporphyrin (CoPP) treatment in arsenical-treated HEK293 cells afforded cytoprotection by attenuating CHOP-associated apoptosis and cytokine mRNA levels. Arsenicals 70-79 heme oxygenase 1 Homo sapiens 8-12 32784000-6 2020 Moreover, CB08035-SCA and CB08035-SYP treatments reduced significantly Bax, BNIP3, APAF1, ERK1, JNK1, MAPK1, NFkappaB1, TNFalpha, IL-6, IL-1beta and HO-1 gene expressions of apoptosis, proinflammation and oxidative stress induced by t-BOOH. SMSF0006723 10-17 heme oxygenase 1 Homo sapiens 149-153 32239675-6 2020 Autophagy inhibitor 3-MA pretreatment reversed the anti-senescent and protective effects on the mitochondrial function of HO-1, which promoted the degradation of the extracellular matrix (ECM) in the intervertebral disc. 3-methyladenine 20-24 heme oxygenase 1 Homo sapiens 122-126 32784000-6 2020 Moreover, CB08035-SCA and CB08035-SYP treatments reduced significantly Bax, BNIP3, APAF1, ERK1, JNK1, MAPK1, NFkappaB1, TNFalpha, IL-6, IL-1beta and HO-1 gene expressions of apoptosis, proinflammation and oxidative stress induced by t-BOOH. SMSF0006723 26-33 heme oxygenase 1 Homo sapiens 149-153 33039895-9 2020 Conversely, genetic or pharmacologic inhibition of HO-1 in TAMs restored the M1-like polarization. tams 59-63 heme oxygenase 1 Homo sapiens 51-55 31983246-0 2020 Thymoquinone and Curcumin combination protects cisplatin-induced Kidney Injury, Nephrotoxicity by attenuating NFkB, KIM-1 and ameliorating Nrf2/HO-1 signaling. Cisplatin 47-56 heme oxygenase 1 Homo sapiens 144-148 31983246-10 2020 Tq + Cur treatment increased the expressions of phosphorylated Akt, Nrf2 and HO-1 proteins while decreasing the levels of cleaved caspase 3 and NFkB in kidney homogenates. thymoquinone 0-2 heme oxygenase 1 Homo sapiens 77-81 31983246-10 2020 Tq + Cur treatment increased the expressions of phosphorylated Akt, Nrf2 and HO-1 proteins while decreasing the levels of cleaved caspase 3 and NFkB in kidney homogenates. Curcumin 5-8 heme oxygenase 1 Homo sapiens 77-81 31983246-11 2020 In summary, Tq + Cur had protective effects on cisplatin-induced nephrotoxicity and renal injury, which could be mediated by up-regulation of survival signals like Akt, Nrf2/HO-1, and attenuation of KIM-1, NFkB. thymoquinone 12-14 heme oxygenase 1 Homo sapiens 174-178 31983246-11 2020 In summary, Tq + Cur had protective effects on cisplatin-induced nephrotoxicity and renal injury, which could be mediated by up-regulation of survival signals like Akt, Nrf2/HO-1, and attenuation of KIM-1, NFkB. Curcumin 17-20 heme oxygenase 1 Homo sapiens 174-178 31983246-11 2020 In summary, Tq + Cur had protective effects on cisplatin-induced nephrotoxicity and renal injury, which could be mediated by up-regulation of survival signals like Akt, Nrf2/HO-1, and attenuation of KIM-1, NFkB. Cisplatin 47-56 heme oxygenase 1 Homo sapiens 174-178 33052744-0 2020 Fisetin Suppresses Pulmonary Inflammatory Responses through Heme Oxygenase-1 Mediated Downregulation of Inducible Nitric Oxide Synthase. fisetin 0-7 heme oxygenase 1 Homo sapiens 60-76 33052744-2 2020 Using lipopolysaccharide (LPS)-activated human pulmonary artery endothelial cells (HPAECs), we determined the effects of fisetin on the induction of heme oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). fisetin 121-128 heme oxygenase 1 Homo sapiens 149-165 33052744-2 2020 Using lipopolysaccharide (LPS)-activated human pulmonary artery endothelial cells (HPAECs), we determined the effects of fisetin on the induction of heme oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2). fisetin 121-128 heme oxygenase 1 Homo sapiens 167-171 32891672-8 2020 Furthermore, we observed that neuroprotective effects of curcumin and riluzole are mediated through Nrf2/HO-1 signaling. Curcumin 57-65 heme oxygenase 1 Homo sapiens 105-109 32891672-8 2020 Furthermore, we observed that neuroprotective effects of curcumin and riluzole are mediated through Nrf2/HO-1 signaling. Riluzole 70-78 heme oxygenase 1 Homo sapiens 105-109 31983246-0 2020 Thymoquinone and Curcumin combination protects cisplatin-induced Kidney Injury, Nephrotoxicity by attenuating NFkB, KIM-1 and ameliorating Nrf2/HO-1 signaling. thymoquinone 0-12 heme oxygenase 1 Homo sapiens 144-148 31983246-0 2020 Thymoquinone and Curcumin combination protects cisplatin-induced Kidney Injury, Nephrotoxicity by attenuating NFkB, KIM-1 and ameliorating Nrf2/HO-1 signaling. Curcumin 17-25 heme oxygenase 1 Homo sapiens 144-148 32716561-7 2020 The potentiation effect of phyllanthin seen with the three ER stressors was related to suppression of survival p-Nrf-2/HO-1 expression, resulting in increased activation of the eIF-2alpha/ATF-4/CHOP pathway. phyllanthin 27-38 heme oxygenase 1 Homo sapiens 119-123 33012373-0 2020 Retraction Notice to "Ketamine Reduces LPS-Induced HMGB1 Through HO-1 and Nrf2/ p38 MAPK" [Journal of Surgical Research 194 (2015) 599-613]. Ketamine 22-30 heme oxygenase 1 Homo sapiens 65-69 33254548-5 2020 HO-1 is induced by hemin, a well-tolerated molecule, used for decades in the treatment of acute intermittent porphyria. Hemin 19-24 heme oxygenase 1 Homo sapiens 0-4 33039895-11 2020 Inhibition of HO-1 overexpression in TAMs may provoke a robust anti-tumor immune response, thereby potentiating the efficacy of chemotherapy. tams 37-41 heme oxygenase 1 Homo sapiens 14-18 32814076-0 2020 Ghrelin mitigates MPP+-induced cytotoxicity: involvement of ERK1/2-mediated Nrf2/HO-1 and endoplasmic reticulum stress PERK signaling pathway. Ghrelin 0-7 heme oxygenase 1 Homo sapiens 81-85 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Biliverdine 110-120 heme oxygenase 1 Homo sapiens 30-35 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. Biliverdine 139-149 heme oxygenase 1 Homo sapiens 29-45 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. Biliverdine 139-149 heme oxygenase 1 Homo sapiens 47-52 32558263-6 2020 Transcription of HMOX1 and other NFE2L2-dependent genes is increased in response to electrophilic and reactive oxygen species (ROS). Reactive Oxygen Species 102-125 heme oxygenase 1 Homo sapiens 17-22 32558263-6 2020 Transcription of HMOX1 and other NFE2L2-dependent genes is increased in response to electrophilic and reactive oxygen species (ROS). Reactive Oxygen Species 127-130 heme oxygenase 1 Homo sapiens 17-22 32558263-9 2020 Transcriptional induction of HMOX1 through derepression of Bach1 with cobalt protoporphyrin (CoPP) or transcriptional activation of HMOX2 by oncogenic B-RafV600E results in increased melanosphere formation. cobaltiprotoporphyrin 70-91 heme oxygenase 1 Homo sapiens 29-34 33000859-0 2020 Madecassoside protects retinal pigment epithelial cells against hydrogen peroxide induced oxidative stress and apoptosis through the activation of Nrf2/HO-1 pathway. madecassoside 0-13 heme oxygenase 1 Homo sapiens 153-157 33000859-10 2020 Furthermore, H2O2-induced increase in expression levels of HO-1 and nuclear Nrf2 were enhanced by MADE treatment. Hydrogen Peroxide 13-17 heme oxygenase 1 Homo sapiens 59-63 33000859-12 2020 In conclusion, these findings demonstrated that MADE protected ARPE-19 cells from H2O2-induced oxidative stress and apoptosis by inducing the activation of Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 82-86 heme oxygenase 1 Homo sapiens 161-165 33110194-5 2020 Growth and differentiation of cells induced by heme-albumin was dependent on heme-oxygenase 1 (HO-1) function and was accompanied with an increase of the intracellular labile iron pool (LIP). Heme 47-51 heme oxygenase 1 Homo sapiens 77-93 33110194-5 2020 Growth and differentiation of cells induced by heme-albumin was dependent on heme-oxygenase 1 (HO-1) function and was accompanied with an increase of the intracellular labile iron pool (LIP). Heme 47-51 heme oxygenase 1 Homo sapiens 95-99 33110194-5 2020 Growth and differentiation of cells induced by heme-albumin was dependent on heme-oxygenase 1 (HO-1) function and was accompanied with an increase of the intracellular labile iron pool (LIP). Iron 175-179 heme oxygenase 1 Homo sapiens 95-99 33079284-6 2022 Additionally, sulforaphane increased the mRNA levels of nuclear factor erythroid-derived 2-like 2 (Nrf2) and heme oxygenase-1 (HO1), both of which mediate several cytoprotective responses. sulforaphane 14-26 heme oxygenase 1 Homo sapiens 109-125 33079284-6 2022 Additionally, sulforaphane increased the mRNA levels of nuclear factor erythroid-derived 2-like 2 (Nrf2) and heme oxygenase-1 (HO1), both of which mediate several cytoprotective responses. sulforaphane 14-26 heme oxygenase 1 Homo sapiens 127-130 33079284-9 2022 Interestingly, the anti-inflammatory and antioxidant effects of sulforaphane were blocked by HO1 pharmacological inhibition, suggesting its dependence on HO1 activity. sulforaphane 64-76 heme oxygenase 1 Homo sapiens 93-96 33079284-9 2022 Interestingly, the anti-inflammatory and antioxidant effects of sulforaphane were blocked by HO1 pharmacological inhibition, suggesting its dependence on HO1 activity. sulforaphane 64-76 heme oxygenase 1 Homo sapiens 154-157 33092263-4 2020 The proline adduct was the most active compound, it showed anti-leukemic activity, upregulated heme oxygenase 1 (HMOX1) and the primary stress-inducible isoform of the heath shock 70 kDa protein 1 (HSPA1A), and downregulated NFkB1 transcription, it was also found to be about 270 times more water soluble than dehydroleucodine. Proline 4-11 heme oxygenase 1 Homo sapiens 95-111 33092263-4 2020 The proline adduct was the most active compound, it showed anti-leukemic activity, upregulated heme oxygenase 1 (HMOX1) and the primary stress-inducible isoform of the heath shock 70 kDa protein 1 (HSPA1A), and downregulated NFkB1 transcription, it was also found to be about 270 times more water soluble than dehydroleucodine. Proline 4-11 heme oxygenase 1 Homo sapiens 113-118 33178028-9 2020 Moreover, UDCA promoted the expression of nuclear Nrf2, HO-1, and NQO1, although arsenic regulated nuclear translocation of Nrf2 positively. Ursodeoxycholic Acid 10-14 heme oxygenase 1 Homo sapiens 56-60 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. Heme 29-33 heme oxygenase 1 Homo sapiens 47-52 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 29-45 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 47-52 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. ammonium ferrous sulfate 130-134 heme oxygenase 1 Homo sapiens 29-45 32558263-4 2020 Two closely related enzymes, heme oxygenase-1 (HMOX1) and heme oxygenase-2 (HMOX2), degrade free heme to produce carbon monoxide, Fe2+ and biliverdin. ammonium ferrous sulfate 130-134 heme oxygenase 1 Homo sapiens 47-52 33142756-8 2020 Furthermore, the mRNA levels of redox-sensitive heme oxygenase (Hmox1), catalase (Cat) and glutamate-cysteine ligase (Gclc) and the total cellular glutathione content were lower in taurine-supplemented cells than they were in the control cells. Taurine 181-188 heme oxygenase 1 Homo sapiens 64-69 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Heme 65-69 heme oxygenase 1 Homo sapiens 12-28 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Heme 65-69 heme oxygenase 1 Homo sapiens 30-35 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 12-28 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 30-35 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. ferrous iron 92-104 heme oxygenase 1 Homo sapiens 12-28 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. ferrous iron 92-104 heme oxygenase 1 Homo sapiens 30-35 33066778-1 2020 BACKGROUND: Heme oxygenase-1 (HMOX1) catalyzes the metabolism of heme into carbon monoxide, ferrous iron, and biliverdin. Biliverdine 110-120 heme oxygenase 1 Homo sapiens 12-28 33076330-3 2020 To identify protection strategies for ADMSCs, this study investigated the influence of the non-psychoactive phytocannabinoid cannabidiol (CBD) and the endocannabinoid analogue R(+)-methanandamide (MA) on the induction of heme oxygenase-1 (HO-1) and autophagy under serum-free conditions. methanandamide 176-195 heme oxygenase 1 Homo sapiens 221-237 33076330-4 2020 At a concentration of 3 microM, CBD induced an upregulation of HO-1 mRNA and protein within 6 h, whereas for MA only a late and comparatively lower increase in the HO-1 protein could be detected after 48 h. In addition, both cannabinoids induced time- and concentration-dependent increases in LC3A/B-II protein, a marker of autophagy, and in metabolic activity. Cannabidiol 32-35 heme oxygenase 1 Homo sapiens 63-67 33076330-6 2020 Similarly, knockdown of HO-1 by siRNA or inhibition of HO-1 activity by tin protoporphyrin IX (SnPPIX) had no effect on CBD-induced autophagy and metabolic activity. tin protoporphyrin IX 72-93 heme oxygenase 1 Homo sapiens 55-59 33076330-6 2020 Similarly, knockdown of HO-1 by siRNA or inhibition of HO-1 activity by tin protoporphyrin IX (SnPPIX) had no effect on CBD-induced autophagy and metabolic activity. tin protoporphyrin IX 95-101 heme oxygenase 1 Homo sapiens 55-59 33076330-9 2020 Remarkably, inhibition of HO-1 by SnPPIX under conditions of autophagy deficit led to a significant reversal of apoptosis in cannabinoid-treated cells. tin protoporphyrin IX 34-40 heme oxygenase 1 Homo sapiens 26-30 33076330-9 2020 Remarkably, inhibition of HO-1 by SnPPIX under conditions of autophagy deficit led to a significant reversal of apoptosis in cannabinoid-treated cells. Cannabinoids 125-136 heme oxygenase 1 Homo sapiens 26-30 33076330-10 2020 In conclusion, the investigated cannabinoids increase metabolic viability of ADMSCs under serum-free conditions by inducing HO-1-independent autophagy but contribute to apoptosis under conditions of additional autophagy deficit via an HO-1-dependent pathway. Cannabinoids 32-44 heme oxygenase 1 Homo sapiens 124-128 33076330-10 2020 In conclusion, the investigated cannabinoids increase metabolic viability of ADMSCs under serum-free conditions by inducing HO-1-independent autophagy but contribute to apoptosis under conditions of additional autophagy deficit via an HO-1-dependent pathway. Cannabinoids 32-44 heme oxygenase 1 Homo sapiens 235-239 33066365-6 2020 Exposure to low O3 concentrations down in the degradation of the explanted adipose tissue and induced a concomitant increase in the protein abundance of Nrf2 and in the expression of its target gene Hmox1. Ozone 16-18 heme oxygenase 1 Homo sapiens 199-204 32860873-3 2020 Endogenously, CO is synthesized upon degradation of heme by heme oxygenases (HOs) of which two enzymatically active isoenzymes are known in mammals; the stress-inducible HO-1 and the constitutively expressed HO-2. Carbon Monoxide 14-16 heme oxygenase 1 Homo sapiens 170-174 32860873-3 2020 Endogenously, CO is synthesized upon degradation of heme by heme oxygenases (HOs) of which two enzymatically active isoenzymes are known in mammals; the stress-inducible HO-1 and the constitutively expressed HO-2. Heme 52-56 heme oxygenase 1 Homo sapiens 170-174 33081396-0 2020 Calcium Ionophore-Induced Extracellular Vesicles Mediate Cytoprotection against Simulated Ischemia/Reperfusion Injury in Cardiomyocyte-Derived Cell Lines by Inducing Heme Oxygenase 1. Calcium 0-7 heme oxygenase 1 Homo sapiens 166-182 33081396-10 2020 Calcium ionophore-induced EVs exert cytoprotection by inducing HO-1 in a TLR4-independent manner. Calcium 0-7 heme oxygenase 1 Homo sapiens 63-67 32860873-4 2020 Among other pathways, HO-1 expression is stimulated by the Nrf2/Keap1 system which senses electrophilic compounds including alkylating agents and reactive oxygen species (ROS) such as superoxide or hydrogen peroxide. Reactive Oxygen Species 146-169 heme oxygenase 1 Homo sapiens 22-26 33053761-7 2020 Geraniol treatment also activated the Keap1/Nrf2/heme oxygenase-1 (HO-1) pathway, increased antioxidant enzyme activities, modulated the PI3K/Akt/mTOR pathway, and ameliorated myocardial autophagy, inflammation, and apoptosis. geraniol 0-8 heme oxygenase 1 Homo sapiens 49-65 32860873-4 2020 Among other pathways, HO-1 expression is stimulated by the Nrf2/Keap1 system which senses electrophilic compounds including alkylating agents and reactive oxygen species (ROS) such as superoxide or hydrogen peroxide. Reactive Oxygen Species 171-174 heme oxygenase 1 Homo sapiens 22-26 32860873-4 2020 Among other pathways, HO-1 expression is stimulated by the Nrf2/Keap1 system which senses electrophilic compounds including alkylating agents and reactive oxygen species (ROS) such as superoxide or hydrogen peroxide. Superoxides 184-194 heme oxygenase 1 Homo sapiens 22-26 32860873-4 2020 Among other pathways, HO-1 expression is stimulated by the Nrf2/Keap1 system which senses electrophilic compounds including alkylating agents and reactive oxygen species (ROS) such as superoxide or hydrogen peroxide. Hydrogen Peroxide 198-215 heme oxygenase 1 Homo sapiens 22-26 32860873-5 2020 In context with ROS, HO-1 expression has been associated with antioxidant defense related to the heme-metabolite redox pair biliverdin/bilirubin. Reactive Oxygen Species 16-19 heme oxygenase 1 Homo sapiens 21-25 32860873-5 2020 In context with ROS, HO-1 expression has been associated with antioxidant defense related to the heme-metabolite redox pair biliverdin/bilirubin. Heme 97-101 heme oxygenase 1 Homo sapiens 21-25 32860873-5 2020 In context with ROS, HO-1 expression has been associated with antioxidant defense related to the heme-metabolite redox pair biliverdin/bilirubin. Biliverdine 124-134 heme oxygenase 1 Homo sapiens 21-25 32860873-5 2020 In context with ROS, HO-1 expression has been associated with antioxidant defense related to the heme-metabolite redox pair biliverdin/bilirubin. Bilirubin 135-144 heme oxygenase 1 Homo sapiens 21-25 32866568-4 2020 Arsenite stimulated an oxidative stress response as detected by Nrf-2 nuclear accumulation and induction of HMOX-1 and NQO1, which was not detected with up to 30 muM uranyl acetate. arsenite 0-8 heme oxygenase 1 Homo sapiens 108-114 33053761-7 2020 Geraniol treatment also activated the Keap1/Nrf2/heme oxygenase-1 (HO-1) pathway, increased antioxidant enzyme activities, modulated the PI3K/Akt/mTOR pathway, and ameliorated myocardial autophagy, inflammation, and apoptosis. geraniol 0-8 heme oxygenase 1 Homo sapiens 67-71 32745467-8 2020 In addition, suppressive effects of gAcrp on ROS production and NADPH oxidase activation, both of which critically contribute to leptin-induced inflammasomes activation, disappeared by inhibition of HO-1 signaling. gacrp 36-41 heme oxygenase 1 Homo sapiens 199-203 33023074-6 2020 Pretreatment with LY294002 (Akt inhibitor) and PD980559 (ERK inhibitor) inhibited LPS-induced Nrf2 nuclear translocation and HO-1 protein expression in ELF-EMF-exposed cells. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 18-26 heme oxygenase 1 Homo sapiens 125-129 33023074-6 2020 Pretreatment with LY294002 (Akt inhibitor) and PD980559 (ERK inhibitor) inhibited LPS-induced Nrf2 nuclear translocation and HO-1 protein expression in ELF-EMF-exposed cells. pd980559 47-55 heme oxygenase 1 Homo sapiens 125-129 32745467-11 2020 In summary, treatment with gAcrp prevents leptin-induced cancer cell growth by modulating inflammasome activation, which is mediated, at least in part, via HO-1 induction and modulation of ER-alpha signaling. gacrp 27-32 heme oxygenase 1 Homo sapiens 156-160 33155244-12 2020 The activity of the NRF2/HO-1 pathway was inhibited by miR-34a-5p, possibly via FOXM1. mir-34a-5p 55-65 heme oxygenase 1 Homo sapiens 25-29 33155244-13 2020 CONCLUSIONS: MiR-34a-5p inhibition attenuates LPS-induced EC injury by targeting FOXM1 via activation of the NRF2/HO-1 pathway. mir-34a-5p 13-23 heme oxygenase 1 Homo sapiens 114-118 32897647-0 2020 miR-141-3p inhibits vascular smooth muscle cell proliferation and migration via regulating Keap1/Nrf2/HO-1 pathway. mir-141-3p 0-10 heme oxygenase 1 Homo sapiens 102-106 32945449-10 2020 Treatment with zinc protoporphyrin-9, a specific inhibitor of heme oxygenase-1 (HO-1), markedly attenuated the cell death induced by MI-463 plus auranofin. zinc protoporphyrin 15-36 heme oxygenase 1 Homo sapiens 62-78 32945449-10 2020 Treatment with zinc protoporphyrin-9, a specific inhibitor of heme oxygenase-1 (HO-1), markedly attenuated the cell death induced by MI-463 plus auranofin. zinc protoporphyrin 15-36 heme oxygenase 1 Homo sapiens 80-84 32945449-10 2020 Treatment with zinc protoporphyrin-9, a specific inhibitor of heme oxygenase-1 (HO-1), markedly attenuated the cell death induced by MI-463 plus auranofin. Auranofin 145-154 heme oxygenase 1 Homo sapiens 62-78 32945449-10 2020 Treatment with zinc protoporphyrin-9, a specific inhibitor of heme oxygenase-1 (HO-1), markedly attenuated the cell death induced by MI-463 plus auranofin. Auranofin 145-154 heme oxygenase 1 Homo sapiens 80-84 32945449-12 2020 In addition, the potent induction of HO-1 contributed to the synergistic effects of MI-463 plus auranofin. Auranofin 96-105 heme oxygenase 1 Homo sapiens 37-41 32897647-11 2020 miR-141-3p also inhibited the expressions of IL-6, IL-beta, TNF-alpha and Keap1 but promoted the expressions of Nrf2 and HO-1. mir-141-3p 0-10 heme oxygenase 1 Homo sapiens 121-125 32897647-13 2020 miR-141-3p is down-regulated during AS, and it can alleviate VSMCs" dysfunction by targeting the Keap1/Nrf2/HO-1 axis. mir-141-3p 0-10 heme oxygenase 1 Homo sapiens 108-112 32712291-6 2020 In addition, hemistepsin A markedly promoted the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was associated with the enhanced expression and activity of heme oxygenase-1 (HO-1) in the presence of 500 muM H2O2. Hydrogen Peroxide 232-236 heme oxygenase 1 Homo sapiens 181-197 32602595-5 2020 HG stimulation-caused the decrease in the expression levels of p-Akt, nuclear Nrf2, and HO-1 was elevated after spiraeoside treatment in AC16 cells. Mercury 0-2 heme oxygenase 1 Homo sapiens 88-92 32602595-5 2020 HG stimulation-caused the decrease in the expression levels of p-Akt, nuclear Nrf2, and HO-1 was elevated after spiraeoside treatment in AC16 cells. spiraeoside 112-123 heme oxygenase 1 Homo sapiens 88-92 32128811-5 2020 We found that BRUP-1 up-regulated the expression level of heme oxygenase-1 (HO-1), one of the rate-limiting enzyme of bilirubin production via nuclear factor erythroid 2-related factor 2 (Nrf2) activation. Bilirubin 118-127 heme oxygenase 1 Homo sapiens 58-74 32128811-5 2020 We found that BRUP-1 up-regulated the expression level of heme oxygenase-1 (HO-1), one of the rate-limiting enzyme of bilirubin production via nuclear factor erythroid 2-related factor 2 (Nrf2) activation. Bilirubin 118-127 heme oxygenase 1 Homo sapiens 76-80 33001278-0 2020 The DPP-IV inhibitor saxagliptin promotes the migration and invasion of papillary thyroid carcinoma cells via the NRF2/HO1 pathway. saxagliptin 21-32 heme oxygenase 1 Homo sapiens 119-122 32707369-19 2020 SCDG medicated serum promoted protein expressions of nuclear Nrf2 and total HO-1, and inhibited translocation of p65. scdg 0-4 heme oxygenase 1 Homo sapiens 76-80 33001278-17 2020 Saxagliptin enhanced the migratory and invasive ability of human thyroid carcinoma cells, as well as the expression of MMP2 and VEGF, by activating the NRF2/HO1 pathway. saxagliptin 0-11 heme oxygenase 1 Homo sapiens 157-160 32712291-6 2020 In addition, hemistepsin A markedly promoted the activation of nuclear factor-erythroid-2-related factor 2 (Nrf2), which was associated with the enhanced expression and activity of heme oxygenase-1 (HO-1) in the presence of 500 muM H2O2. Hydrogen Peroxide 232-236 heme oxygenase 1 Homo sapiens 199-203 33134292-0 2020 Celecoxib Exerts Neuroprotective Effects in beta-Amyloid-Treated SH-SY5Y Cells Through the Regulation of Heme Oxygenase-1: Novel Insights for an Old Drug. Celecoxib 0-9 heme oxygenase 1 Homo sapiens 105-121 33134292-5 2020 Both sAbeta (6.25-50 nM) and fAbeta (1.25-50 nM) dose-dependently over-expressed inducible HO (HO-1) after 24 h of incubation, reaching statistical significance at 25 and 6.25 nM, respectively. sabeta 5-11 heme oxygenase 1 Homo sapiens 95-99 32993497-3 2020 H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Deuterium 0-3 heme oxygenase 1 Homo sapiens 106-110 32993497-3 2020 H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Carbon Monoxide 28-43 heme oxygenase 1 Homo sapiens 106-110 32993497-3 2020 H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Carbon Monoxide 45-47 heme oxygenase 1 Homo sapiens 106-110 32993497-3 2020 H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Heme 72-76 heme oxygenase 1 Homo sapiens 106-110 32993497-3 2020 H2S release is triggered by carbon monoxide (CO) from the catabolism of heme by inducible heme oxygenase (HO-1) and heme proteins possess catalytic activity necessary for the H2S signalling by protein persulfidation. Deuterium 175-178 heme oxygenase 1 Homo sapiens 106-110 32993497-9 2020 Finally, an array of therapeutic interventions with the potential to clinically modulate the HO-1/CO/H2S axis is already available or under development. Carbon Monoxide 98-100 heme oxygenase 1 Homo sapiens 93-97 32993497-9 2020 Finally, an array of therapeutic interventions with the potential to clinically modulate the HO-1/CO/H2S axis is already available or under development. Deuterium 101-104 heme oxygenase 1 Homo sapiens 93-97 33134292-5 2020 Both sAbeta (6.25-50 nM) and fAbeta (1.25-50 nM) dose-dependently over-expressed inducible HO (HO-1) after 24 h of incubation, reaching statistical significance at 25 and 6.25 nM, respectively. fabeta 29-35 heme oxygenase 1 Homo sapiens 95-99 33134292-7 2020 Furthermore, 10 muM CXB counteracted the Abeta-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 muM CXB significantly counteracted only 25 nM sAbeta-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. Celecoxib 20-23 heme oxygenase 1 Homo sapiens 106-110 33134292-7 2020 Furthermore, 10 muM CXB counteracted the Abeta-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 muM CXB significantly counteracted only 25 nM sAbeta-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. Celecoxib 20-23 heme oxygenase 1 Homo sapiens 263-267 33134292-7 2020 Furthermore, 10 muM CXB counteracted the Abeta-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 muM CXB significantly counteracted only 25 nM sAbeta-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. Celecoxib 147-150 heme oxygenase 1 Homo sapiens 263-267 33134292-7 2020 Furthermore, 10 muM CXB counteracted the Abeta-induced ROS production with a mechanism fully dependent on HO-1 up-regulation; nevertheless, 10 muM CXB significantly counteracted only 25 nM sAbeta-induced lipid peroxidation damage in SH-SY5Y neurons by modulating HO-1. sabeta 189-195 heme oxygenase 1 Homo sapiens 263-267 32968051-0 2020 Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Reactive Oxygen Species 52-55 heme oxygenase 1 Homo sapiens 0-16 32992548-9 2020 In conclusion, MG inhibited hepatic steatosis-induced NLRP3 inflammasome activation through the restoration of autophagy to promote HO-1 signaling capable of ameliorating oxidative stress and inflammatory responses. magnolol 15-17 heme oxygenase 1 Homo sapiens 132-136 32992548-0 2020 Involvement of HO-1 and Autophagy in the Protective Effect of Magnolol in Hepatic Steatosis-Induced NLRP3 Inflammasome Activation In Vivo and In Vitro. magnolol 62-70 heme oxygenase 1 Homo sapiens 15-19 32968051-11 2020 Chemical inhibition or genetic knockdown of HO-1 potentiated hemin-triggered ROS generation and oxidative DNA damage preferentially in HCEC. Hemin 61-66 heme oxygenase 1 Homo sapiens 44-48 32968051-0 2020 Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Heme 116-120 heme oxygenase 1 Homo sapiens 0-16 32968051-11 2020 Chemical inhibition or genetic knockdown of HO-1 potentiated hemin-triggered ROS generation and oxidative DNA damage preferentially in HCEC. Reactive Oxygen Species 77-80 heme oxygenase 1 Homo sapiens 44-48 32968051-0 2020 Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron. Iron 121-125 heme oxygenase 1 Homo sapiens 0-16 32968051-12 2020 Furthermore, HO-1 abrogation strongly augmented the cytotoxic effects of hemin in HCEC, revealing its pivotal function in colonocytes and highlighting the toxicity of free intracellular heme iron. Hemin 73-78 heme oxygenase 1 Homo sapiens 13-17 32968051-12 2020 Furthermore, HO-1 abrogation strongly augmented the cytotoxic effects of hemin in HCEC, revealing its pivotal function in colonocytes and highlighting the toxicity of free intracellular heme iron. Heme 186-190 heme oxygenase 1 Homo sapiens 13-17 32968051-9 2020 Hemin caused stabilization and nuclear translocation of Nrf2, which induced heme oxygenase-1 (HO-1) and ferritin heavy chain (FtH). Hemin 0-5 heme oxygenase 1 Homo sapiens 76-92 32968051-12 2020 Furthermore, HO-1 abrogation strongly augmented the cytotoxic effects of hemin in HCEC, revealing its pivotal function in colonocytes and highlighting the toxicity of free intracellular heme iron. Iron 191-195 heme oxygenase 1 Homo sapiens 13-17 32968051-9 2020 Hemin caused stabilization and nuclear translocation of Nrf2, which induced heme oxygenase-1 (HO-1) and ferritin heavy chain (FtH). Hemin 0-5 heme oxygenase 1 Homo sapiens 94-98 32968051-14 2020 Importantly, HO-1 conferred protection against the detrimental effects of hemin. Hemin 74-79 heme oxygenase 1 Homo sapiens 13-17 32967203-12 2020 Our results indicate that piperine may be an effective molecule for the prophylactic treatment of colon carcinogenesis by targeting the NF-kappaB/Nrf-2/Keap-1/HO-1 pathway as a progressive strategy in the preclusion and effective treatment of colorectal cancer. piperine 26-34 heme oxygenase 1 Homo sapiens 159-163 32971746-1 2020 Heme oxygenase-1 is induced by many cellular stressors and catalyzes the breakdown of heme to generate carbon monoxide and bilirubin, which confer cytoprotection. Heme 86-90 heme oxygenase 1 Homo sapiens 0-16 32971746-1 2020 Heme oxygenase-1 is induced by many cellular stressors and catalyzes the breakdown of heme to generate carbon monoxide and bilirubin, which confer cytoprotection. Carbon Monoxide 103-118 heme oxygenase 1 Homo sapiens 0-16 32971746-1 2020 Heme oxygenase-1 is induced by many cellular stressors and catalyzes the breakdown of heme to generate carbon monoxide and bilirubin, which confer cytoprotection. Bilirubin 123-132 heme oxygenase 1 Homo sapiens 0-16 32971746-8 2020 Our results suggest that regulation of heme may be an equally significant role of HO-1. Heme 39-43 heme oxygenase 1 Homo sapiens 82-86 32967203-9 2020 We found that piperine inhibited NF-kappaB by the activation of Nrf-2, blocking downstream inflammatory mediators/cytokines (TNF-alpha, IL-6, IL-1beta, Cox-2, PGE-2, iNOS, NO, MPO), triggering an antioxidant response machinery (HO-1, NQO-1, GSH, GR, GPx, CAT, SOD), scavenging ROS, and decreasing lipid peroxidation. piperine 14-22 heme oxygenase 1 Homo sapiens 228-232 32553625-8 2020 We also found that DMF inhibited the extracellular release of high mobility group box 1, associated with an up-regulation of heme oxygenase-1, likely mediated through Nrf2 activation. Dimethyl Fumarate 19-22 heme oxygenase 1 Homo sapiens 125-141 32615411-0 2020 Nrf2/HO-1 partially regulates cytoprotective effects of carbon monoxide against urban particulate matter-induced inflammatory responses in oral keratinocytes. Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 5-9 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Heme 73-77 heme oxygenase 1 Homo sapiens 0-16 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Heme 73-77 heme oxygenase 1 Homo sapiens 18-22 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Biliverdine 81-91 heme oxygenase 1 Homo sapiens 0-16 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Biliverdine 81-91 heme oxygenase 1 Homo sapiens 18-22 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 0-16 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 18-22 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. ferrous iron 118-130 heme oxygenase 1 Homo sapiens 0-16 32880973-1 2021 Heme oxygenase-1 (HO-1), a highly inducible stress protein that degrades heme to biliverdin, carbon monoxide and free ferrous iron, is increased in blood and other biofluids of subjects with various systemic and neurological disorders. ferrous iron 118-130 heme oxygenase 1 Homo sapiens 18-22 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Heme 31-35 heme oxygenase 1 Homo sapiens 208-224 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Heme 31-35 heme oxygenase 1 Homo sapiens 226-231 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Heme 31-35 heme oxygenase 1 Homo sapiens 233-237 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Adenosine 49-58 heme oxygenase 1 Homo sapiens 208-224 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Adenosine 49-58 heme oxygenase 1 Homo sapiens 226-231 32611235-2 2020 In human macrophages in vitro, heme activates an adenosine monophosphate activated protein kinase / activating transcription factor 1 (AMPK/ATF1) pathway that directs Mhem macrophages through coregulation of heme oxygenase 1 (HMOX1, HO-1) and lipid homeostasis genes. Adenosine 49-58 heme oxygenase 1 Homo sapiens 233-237 32913257-5 2020 Methylglyoxal treatment also significantly decreased the mRNA expression of the slit diaphragm markers ZO-1 and NEPH1 and significantly increased the mRNA expression of the oxidative stress marker HO-1. Pyruvaldehyde 0-13 heme oxygenase 1 Homo sapiens 197-201 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Carbon Monoxide 63-78 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Carbon Monoxide 63-78 heme oxygenase 1 Homo sapiens 18-22 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Carbon Monoxide 80-82 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Carbon Monoxide 80-82 heme oxygenase 1 Homo sapiens 18-22 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Iron 85-89 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Iron 85-89 heme oxygenase 1 Homo sapiens 18-22 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 18-22 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Bilirubin 139-148 heme oxygenase 1 Homo sapiens 0-16 32899732-1 2020 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme into carbon monoxide (CO), iron, and biliverdin, which is rapidly metabolized to bilirubin. Bilirubin 139-148 heme oxygenase 1 Homo sapiens 18-22 32899732-4 2020 The vasoprotection evoked by HO-1 is largely ascribed to the generation of CO and/or the bile pigments, biliverdin and bilirubin, which exert potent antioxidant and anti-inflammatory effects. Carbon Monoxide 75-77 heme oxygenase 1 Homo sapiens 29-33 32899732-4 2020 The vasoprotection evoked by HO-1 is largely ascribed to the generation of CO and/or the bile pigments, biliverdin and bilirubin, which exert potent antioxidant and anti-inflammatory effects. Biliverdine 104-114 heme oxygenase 1 Homo sapiens 29-33 32899732-4 2020 The vasoprotection evoked by HO-1 is largely ascribed to the generation of CO and/or the bile pigments, biliverdin and bilirubin, which exert potent antioxidant and anti-inflammatory effects. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 29-33 32899732-6 2020 Several therapeutic strategies are currently being pursued that may allow for the targeting of HO-1 in arterial remodeling in various pathologies, including the use of gene delivery approaches, the development of novel inducers of the enzyme, and the administration of unique formulations of CO and bilirubin. Carbon Monoxide 292-294 heme oxygenase 1 Homo sapiens 95-99 32899732-6 2020 Several therapeutic strategies are currently being pursued that may allow for the targeting of HO-1 in arterial remodeling in various pathologies, including the use of gene delivery approaches, the development of novel inducers of the enzyme, and the administration of unique formulations of CO and bilirubin. Bilirubin 299-308 heme oxygenase 1 Homo sapiens 95-99 32574971-7 2020 The underlying mechanisms of monotropein on alleviating cisplatin-induced AKI were associated with the activation of Nrf2/HO-1 pathway against oxidative stress and the inhibition on NF-kappaB signaling to suppress inflammation as well as the regulation on the expressions of proteins in apoptosis pathway in this renal injury model. Cisplatin 56-65 heme oxygenase 1 Homo sapiens 122-126 32500379-3 2020 Additionally, COVID-19"s heme reduction may contribute to even lower HO-1. Heme 25-29 heme oxygenase 1 Homo sapiens 69-73 32500379-9 2020 Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Sevoflurane 82-93 heme oxygenase 1 Homo sapiens 48-52 32500379-9 2020 Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Isoflurane 97-107 heme oxygenase 1 Homo sapiens 48-52 32500379-9 2020 Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Curcumin 136-144 heme oxygenase 1 Homo sapiens 48-52 32500379-9 2020 Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Resveratrol 146-157 heme oxygenase 1 Homo sapiens 48-52 32500379-9 2020 Therapies with anti-viral properties that raise HO-1 include certain anesthetics (sevoflurane or isoflurane), hemin, estrogen, statins, curcumin, resveratrol, and melatonin. Melatonin 163-172 heme oxygenase 1 Homo sapiens 48-52 32293791-0 2020 Emodin protected against synaptic impairment and oxidative stress induced by fluoride in SH-SY5Y cells by modulating ERK1/2/Nrf2/HO-1 pathway. Emodin 0-6 heme oxygenase 1 Homo sapiens 129-133 32293791-0 2020 Emodin protected against synaptic impairment and oxidative stress induced by fluoride in SH-SY5Y cells by modulating ERK1/2/Nrf2/HO-1 pathway. Fluorides 77-85 heme oxygenase 1 Homo sapiens 129-133 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 78-94 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 96-100 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 78-94 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 96-100 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Heme 34-38 heme oxygenase 1 Homo sapiens 78-94 32615411-3 2020 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by heme oxygenase-1 (HO-1), has been shown to own cytoprotective effects including anti-inflammation and antioxidant. Heme 34-38 heme oxygenase 1 Homo sapiens 96-100 32347305-11 2020 OUTCOMES: HO-1 exerts some protective effects on the placentation, probably by a combination of factors, including its interrelation with the PGC-1alpha/PPAR pathway and the sFlt1/PlGF balance, and through its primary metabolites, notably carbon monoxide and bilirubin. Carbon Monoxide 239-254 heme oxygenase 1 Homo sapiens 10-14 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Carbon Dioxide 141-143 heme oxygenase 1 Homo sapiens 47-63 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Carbon Dioxide 141-143 heme oxygenase 1 Homo sapiens 65-69 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Biliverdine 145-155 heme oxygenase 1 Homo sapiens 47-63 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Biliverdine 145-155 heme oxygenase 1 Homo sapiens 65-69 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Bilirubin 160-169 heme oxygenase 1 Homo sapiens 47-63 31830562-4 2020 Activation of Nrf2 results in the synthesis of heme oxygenase-1 (HO-1) leading to the formation of a number of bioactive metabolites, mainly CO, biliverdin and bilirubin. Bilirubin 160-169 heme oxygenase 1 Homo sapiens 65-69 32347305-11 2020 OUTCOMES: HO-1 exerts some protective effects on the placentation, probably by a combination of factors, including its interrelation with the PGC-1alpha/PPAR pathway and the sFlt1/PlGF balance, and through its primary metabolites, notably carbon monoxide and bilirubin. Bilirubin 259-268 heme oxygenase 1 Homo sapiens 10-14 32940965-1 2020 BACKGROUND: Tin protoporphyrin (SnPP), a heme oxygenase 1 (HO-1) inhibitor, triggers adaptive tissue responses that confer potent protection against acute renal- and extra-renal tissue injuries. tin protoporphyrin IX 12-30 heme oxygenase 1 Homo sapiens 41-57 32540488-9 2020 Moreover, the nuclear factor erythoid 2-related 2 (Nrf2) and HO-1 expression were lower in 3% DSS+ Ferr-1 group than 3% DSS group (P < 0.05). Dextran Sulfate 94-97 heme oxygenase 1 Homo sapiens 61-65 32540488-10 2020 These data revealed that suppressing ferroptosis could effectively ameliorate DSS-induced UC involved in blocking Nrf2/HO-1 signaling pathway. Dextran Sulfate 78-81 heme oxygenase 1 Homo sapiens 119-123 32782567-0 2020 Enhancement of piperlongumine chemosensitivity by silencing heme oxygenase-1 expression in cholangiocarcinoma cell lines. piperlonguminine 15-29 heme oxygenase 1 Homo sapiens 60-76 32782567-3 2020 The aim of the present study was to investigate the molecular mechanisms of PL-induced heme oxygenase-1 (HO-1) expression in CCA cell lines. piperlonguminine 76-78 heme oxygenase 1 Homo sapiens 87-103 32782567-3 2020 The aim of the present study was to investigate the molecular mechanisms of PL-induced heme oxygenase-1 (HO-1) expression in CCA cell lines. piperlonguminine 76-78 heme oxygenase 1 Homo sapiens 105-109 32782567-10 2020 Dose-dependent upregulation of HO-1 expression via PI3K/Akt activation was detected in PL-treated CCA cells. piperlonguminine 87-89 heme oxygenase 1 Homo sapiens 31-35 32782567-11 2020 Inhibition of HO-1 eliminated the antioxidant defense mechanisms, leading to increased anti-cancer activity of PL in the CCA cell lines via an increase in intracellular ROS levels and apoptotic protein expression. piperlonguminine 111-113 heme oxygenase 1 Homo sapiens 14-18 32782567-11 2020 Inhibition of HO-1 eliminated the antioxidant defense mechanisms, leading to increased anti-cancer activity of PL in the CCA cell lines via an increase in intracellular ROS levels and apoptotic protein expression. Reactive Oxygen Species 169-172 heme oxygenase 1 Homo sapiens 14-18 32782567-12 2020 These observations indicated that HO-1 inhibition had a chemosensitizing effect on CCA to PL. piperlonguminine 90-92 heme oxygenase 1 Homo sapiens 34-38 32540488-0 2020 Ferroptosis mediated DSS-induced ulcerative colitis associated with Nrf2/HO-1 signaling pathway. Dextran Sulfate 21-24 heme oxygenase 1 Homo sapiens 73-77 32645628-0 2020 DL0410 attenuates oxidative stress and neuroinflammation via BDNF/TrkB/ERK/CREB and Nrf2/HO-1 activation. DL0410 0-6 heme oxygenase 1 Homo sapiens 89-93 32940965-1 2020 BACKGROUND: Tin protoporphyrin (SnPP), a heme oxygenase 1 (HO-1) inhibitor, triggers adaptive tissue responses that confer potent protection against acute renal- and extra-renal tissue injuries. tin protoporphyrin IX 12-30 heme oxygenase 1 Homo sapiens 59-63 32940965-1 2020 BACKGROUND: Tin protoporphyrin (SnPP), a heme oxygenase 1 (HO-1) inhibitor, triggers adaptive tissue responses that confer potent protection against acute renal- and extra-renal tissue injuries. S-Nitroso-N-propionyl-D,L-penicillamine 32-36 heme oxygenase 1 Homo sapiens 41-57 32940965-1 2020 BACKGROUND: Tin protoporphyrin (SnPP), a heme oxygenase 1 (HO-1) inhibitor, triggers adaptive tissue responses that confer potent protection against acute renal- and extra-renal tissue injuries. S-Nitroso-N-propionyl-D,L-penicillamine 32-36 heme oxygenase 1 Homo sapiens 59-63 32150044-12 2020 Dual inhibition of both miR-24-3p and miR-145-5p prior to hypoxia-reoxygenation caused significant upregulation of SOD2 and HMOX1 protein; fold-change of 3.17 (p<=0.05) and 6.97 (p<=0.05) respectively. mir-24-3p 24-33 heme oxygenase 1 Homo sapiens 124-129 32506039-4 2020 In keratinocytes, DHA-exposure performed at low millimolar concentrations did not impair viability while causing a pronounced cellular stress response as obvious from rapid activation of phospho-protein signal transduction [p-p38, p-Hsp27(S15/S78), p-eIF2alpha] and gene expression changes (HSPA6, HMOX1, CRYAB, CCL3), not observable upon exposure to the non-ketose, tanning-inactive DHA-control glycerol. Dihydroxyacetone 18-21 heme oxygenase 1 Homo sapiens 298-303 32506039-6 2020 In human epidermal reconstructs a topical use-relevant DHA-dose regimen elicited a comparable stress response as revealed by gene expression array (HSPA1A, HSPA6, HSPD1, IL6, DDIT3, EGR1) and immunohistochemical analysis (CEL, HO-1, p-Hsp27-S78). Dihydroxyacetone 55-58 heme oxygenase 1 Homo sapiens 227-231 32828015-6 2020 In addition, the leukocytes from people with HIV with CWP had attenuated levels of the heme metabolizing enzyme, heme oxygenase-1, which metabolizes free heme to carbon-monoxide and biliverdin. Heme 87-91 heme oxygenase 1 Homo sapiens 113-129 32828015-6 2020 In addition, the leukocytes from people with HIV with CWP had attenuated levels of the heme metabolizing enzyme, heme oxygenase-1, which metabolizes free heme to carbon-monoxide and biliverdin. Carbon Monoxide 162-177 heme oxygenase 1 Homo sapiens 113-129 32828015-6 2020 In addition, the leukocytes from people with HIV with CWP had attenuated levels of the heme metabolizing enzyme, heme oxygenase-1, which metabolizes free heme to carbon-monoxide and biliverdin. Biliverdine 182-192 heme oxygenase 1 Homo sapiens 113-129 32982786-5 2020 HO-1 and HO-2 catalyze the rate-limiting step of cellular heme degradation and, similar to SirT1, HO-1 exerts beneficial effects in the vasculature through the activation of anti-oxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative signaling pathways. Heme 58-62 heme oxygenase 1 Homo sapiens 0-4 32982732-0 2020 Jian-Pi-Bu-Xue-Formula Alleviates Cyclophosphamide-Induced Myelosuppression via Up-Regulating NRF2/HO1/NQO1 Signaling. jian-pi-bu-xue 0-14 heme oxygenase 1 Homo sapiens 99-102 32982732-0 2020 Jian-Pi-Bu-Xue-Formula Alleviates Cyclophosphamide-Induced Myelosuppression via Up-Regulating NRF2/HO1/NQO1 Signaling. Cyclophosphamide 34-50 heme oxygenase 1 Homo sapiens 99-102 32854434-0 2020 Cudratricusxanthone O Inhibits H2O2-Induced Cell Damage by Activating Nrf2/HO-1 Pathway in Human Chondrocytes. cudratricusxanthone 0-19 heme oxygenase 1 Homo sapiens 75-79 32854434-0 2020 Cudratricusxanthone O Inhibits H2O2-Induced Cell Damage by Activating Nrf2/HO-1 Pathway in Human Chondrocytes. Hydrogen Peroxide 31-35 heme oxygenase 1 Homo sapiens 75-79 32854434-2 2020 Although induced-heme oxygenase-1 (HO-1) has been found to protect cells against oxygen radical damage, little information is available regarding the use of bioactive compounds from natural sources for regulating the HO-1 pathway to treat OA. Oxygen 22-28 heme oxygenase 1 Homo sapiens 35-39 32854434-3 2020 In this study, we explored the inhibitory effects of cudratricusxanthone O (CTO) isolated from the Maclura tricuspidata Bureau (Moraceae) on H2O2-induced damage of SW1353 chondrocytes via regulation of the HO-1 pathway. cudratricusxanthone o 53-74 heme oxygenase 1 Homo sapiens 206-210 32854434-3 2020 In this study, we explored the inhibitory effects of cudratricusxanthone O (CTO) isolated from the Maclura tricuspidata Bureau (Moraceae) on H2O2-induced damage of SW1353 chondrocytes via regulation of the HO-1 pathway. cto 76-79 heme oxygenase 1 Homo sapiens 206-210 32854434-7 2020 Treatment with the HO-1 inhibitor tin-protoporphyrin IX revealed that these protective effects were exerted due to an increase in HO-1 expression induced by CTO. tin protoporphyrin IX 34-55 heme oxygenase 1 Homo sapiens 19-23 32908872-0 2020 Retracted: Involvement of Nrf2-Mediated Upregulation of Heme Oxygenase-1 in Mollugin-Induced Growth Inhibition and Apoptosis in Human Oral Cancer Cells. rubimaillin 76-84 heme oxygenase 1 Homo sapiens 56-72 32854434-7 2020 Treatment with the HO-1 inhibitor tin-protoporphyrin IX revealed that these protective effects were exerted due to an increase in HO-1 expression induced by CTO. tin protoporphyrin IX 34-55 heme oxygenase 1 Homo sapiens 130-134 32854434-7 2020 Treatment with the HO-1 inhibitor tin-protoporphyrin IX revealed that these protective effects were exerted due to an increase in HO-1 expression induced by CTO. cto 157-160 heme oxygenase 1 Homo sapiens 19-23 32854434-7 2020 Treatment with the HO-1 inhibitor tin-protoporphyrin IX revealed that these protective effects were exerted due to an increase in HO-1 expression induced by CTO. cto 157-160 heme oxygenase 1 Homo sapiens 130-134 32842569-7 2020 Besides, BA downregulated the phosphorylation of the p38, JNK, and ERK proteins, while it upregulated the expression of the Nrf2 and HO-1 proteins in thymus tissues. betulinic acid 9-11 heme oxygenase 1 Homo sapiens 133-137 32908636-6 2020 HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Carbon Monoxide 37-52 heme oxygenase 1 Homo sapiens 0-4 32908636-6 2020 HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Carbon Monoxide 54-56 heme oxygenase 1 Homo sapiens 0-4 32908636-6 2020 HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Iron 64-68 heme oxygenase 1 Homo sapiens 0-4 32908636-6 2020 HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Biliverdine 78-88 heme oxygenase 1 Homo sapiens 0-4 32908636-6 2020 HO-1 activity leads to production of carbon monoxide (CO), free iron ion, and biliverdin; the latter is promptly reduced to bilirubin. Bilirubin 124-133 heme oxygenase 1 Homo sapiens 0-4 32913552-5 2020 This study was designed to examine the role of HO-1 in lead acetate (PbAc)-induced renal tubular cell injury in vitro. lead acetate 55-67 heme oxygenase 1 Homo sapiens 47-51 32818234-11 2020 RSG cotreatment significantly protected against HQ-induced necrosis and apoptosis, prevented HQ-reduced mitochondrial bioenergetics, decreased HQ-induced reactive oxygen species production, improved HQ-disrupted mitochondrial membrane potential, reduced F-actin aggregates, decreased phosphorylation of P38 and heat shock protein 27, and further upregulated HQ-induced heme oxygenase-1 protein levels. rsg 0-3 heme oxygenase 1 Homo sapiens 369-385 32913552-5 2020 This study was designed to examine the role of HO-1 in lead acetate (PbAc)-induced renal tubular cell injury in vitro. pbac 69-73 heme oxygenase 1 Homo sapiens 47-51 32913552-8 2020 In addition, pretreatment with 3-methyladenine, an inhibitor of autophagy, aggravated apoptosis and abolished renoprotection by HO-1, suggesting that the anti-apoptotic effect of HO-1 in Pb-induced nephrotoxicity is dependent on enhanced autophagy. 3-methyladenine 31-46 heme oxygenase 1 Homo sapiens 128-132 32913552-8 2020 In addition, pretreatment with 3-methyladenine, an inhibitor of autophagy, aggravated apoptosis and abolished renoprotection by HO-1, suggesting that the anti-apoptotic effect of HO-1 in Pb-induced nephrotoxicity is dependent on enhanced autophagy. 3-methyladenine 31-46 heme oxygenase 1 Homo sapiens 179-183 32913552-8 2020 In addition, pretreatment with 3-methyladenine, an inhibitor of autophagy, aggravated apoptosis and abolished renoprotection by HO-1, suggesting that the anti-apoptotic effect of HO-1 in Pb-induced nephrotoxicity is dependent on enhanced autophagy. Lead 187-189 heme oxygenase 1 Homo sapiens 179-183 32913552-12 2020 Our findings suggest that HO-1 alleviates Pb-induced nephrotoxicity via enhanced autophagy, which involves activation of the AMPK/mTORC1 signaling pathway. Lead 42-44 heme oxygenase 1 Homo sapiens 26-30 32801826-9 2020 We established a central role for L-cystine, as changes in the expression of the anti-oxidative gene hmox1 were L-cystine-dependent. Cystine 34-43 heme oxygenase 1 Homo sapiens 101-106 32801826-9 2020 We established a central role for L-cystine, as changes in the expression of the anti-oxidative gene hmox1 were L-cystine-dependent. Cystine 112-121 heme oxygenase 1 Homo sapiens 101-106 32817372-7 2020 In parallel with decreased GULP1 expression, we observed increased expression of NRF2, HMOX1, and other candidate antioxidant genes in cisplatin-resistant cells. Cisplatin 135-144 heme oxygenase 1 Homo sapiens 87-92 32416241-3 2020 The present work tested the hypothesis that zinc is necessary in the neuronal defense response against dopamine (DA)-induced oxidative stress, in particular through heme oxygenase-1 (HO-1) upregulation. Dopamine 103-111 heme oxygenase 1 Homo sapiens 183-187 32416241-3 2020 The present work tested the hypothesis that zinc is necessary in the neuronal defense response against dopamine (DA)-induced oxidative stress, in particular through heme oxygenase-1 (HO-1) upregulation. Dopamine 113-115 heme oxygenase 1 Homo sapiens 165-181 32416241-3 2020 The present work tested the hypothesis that zinc is necessary in the neuronal defense response against dopamine (DA)-induced oxidative stress, in particular through heme oxygenase-1 (HO-1) upregulation. Dopamine 113-115 heme oxygenase 1 Homo sapiens 183-187 32416241-5 2020 Human IMR-32 neuroblastoma cells responded to high DA concentrations (100 muM) by upregulating HO-1. Dopamine 51-53 heme oxygenase 1 Homo sapiens 95-99 32416241-7 2020 DA-mediated induction of HO-1 expression was dependent on the concentration of zinc in the medium. Dopamine 0-2 heme oxygenase 1 Homo sapiens 25-29 32416241-12 2020 Results suggest that zinc is crucial in the neuronal response to DA-induced oxidative stress in part through its role in the modulation of the Nrf2-and Bach-1-driven upregulation of HO-1 expression. Dopamine 65-67 heme oxygenase 1 Homo sapiens 182-186 32567157-8 2020 ATX and LiCl treatment raised the protein levels of p-Akt, p-GSK3beta, nucleus Nrf2 and haeme oxygenase 1 (HO-1). astaxanthine 0-3 heme oxygenase 1 Homo sapiens 88-105 32567157-8 2020 ATX and LiCl treatment raised the protein levels of p-Akt, p-GSK3beta, nucleus Nrf2 and haeme oxygenase 1 (HO-1). astaxanthine 0-3 heme oxygenase 1 Homo sapiens 107-111 32567157-8 2020 ATX and LiCl treatment raised the protein levels of p-Akt, p-GSK3beta, nucleus Nrf2 and haeme oxygenase 1 (HO-1). Lithium Chloride 8-12 heme oxygenase 1 Homo sapiens 88-105 32567157-8 2020 ATX and LiCl treatment raised the protein levels of p-Akt, p-GSK3beta, nucleus Nrf2 and haeme oxygenase 1 (HO-1). Lithium Chloride 8-12 heme oxygenase 1 Homo sapiens 107-111 32559798-1 2020 OBJECTIVE: We examined the interaction of polymorphisms in the genes heme oxygenase-1 (HMOX1) and nitric oxide synthase (NOS3) in patients with preeclampsia (PE) as well as the responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 196-206 heme oxygenase 1 Homo sapiens 88-93 32407842-0 2020 Excessive reactive oxygen species induce apoptosis via the APPL1-Nrf2/HO-1 antioxidant signalling pathway in trophoblasts with missed abortion. Reactive Oxygen Species 10-33 heme oxygenase 1 Homo sapiens 70-74 31833548-8 2020 In Vitro assay revealed that mutant IDH indirectly reduced the amount of exogenous 5-ALA-derived protoporphyrinogen IX in glioma cells by increasing activity of ferrochelatase and heme oxygenase 1. Aminolevulinic Acid 83-88 heme oxygenase 1 Homo sapiens 180-196 32155298-0 2020 Ampelopsin inhibits high glucose-induced extracellular matrix accumulation and oxidative stress in mesangial cells through activating the Nrf2/HO-1 pathway. Glucose 25-32 heme oxygenase 1 Homo sapiens 143-147 32155298-9 2020 Furthermore, AMP elevated the expression levels of nuclear Nrf2 and heme oxygenase-1 (HO-1), as well as increased the mRNA levels of Nrf2-driven genes NAD(P)H dehydrogenase quinone-1 (NQO-1) and HO-1 in HG-treated MCs. ampelopsin 13-16 heme oxygenase 1 Homo sapiens 68-84 32155298-9 2020 Furthermore, AMP elevated the expression levels of nuclear Nrf2 and heme oxygenase-1 (HO-1), as well as increased the mRNA levels of Nrf2-driven genes NAD(P)H dehydrogenase quinone-1 (NQO-1) and HO-1 in HG-treated MCs. ampelopsin 13-16 heme oxygenase 1 Homo sapiens 86-90 32155298-9 2020 Furthermore, AMP elevated the expression levels of nuclear Nrf2 and heme oxygenase-1 (HO-1), as well as increased the mRNA levels of Nrf2-driven genes NAD(P)H dehydrogenase quinone-1 (NQO-1) and HO-1 in HG-treated MCs. ampelopsin 13-16 heme oxygenase 1 Homo sapiens 195-199 32155298-11 2020 In conclusion, these findings indicated that AMP protected MCs from HG-induced oxidative damage and ECM accumulation, which might be mediated by Nrf2/HO-1 pathway. ampelopsin 45-48 heme oxygenase 1 Homo sapiens 150-154 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 91-101 heme oxygenase 1 Homo sapiens 47-53 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 91-101 heme oxygenase 1 Homo sapiens 115-120 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 91-101 heme oxygenase 1 Homo sapiens 163-167 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 189-199 heme oxygenase 1 Homo sapiens 47-53 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 189-199 heme oxygenase 1 Homo sapiens 115-120 32559798-3 2020 RESULTS: We found interactions between genotypes of the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that PE genotyped as AT presents lower levels of protein HO-1 compared with AA. Methyldopa 101-111 heme oxygenase 1 Homo sapiens 57-63 32559798-4 2020 CONCLUSION: We found interactions between the HMOX-1 and NOS3 genes and responsiveness to methyldopa and that the HMOX1 polymorphism affects the levels of enzyme HO-1 in responsiveness to methyldopa and to total antihypertensive therapy. Methyldopa 189-199 heme oxygenase 1 Homo sapiens 163-167 32850957-0 2020 Melatonin Inhibits Oxidative Stress and Apoptosis in Cryopreserved Ovarian Tissues via Nrf2/HO-1 Signaling Pathway. Melatonin 0-9 heme oxygenase 1 Homo sapiens 92-96 32737955-7 2020 We identified the levels of expression of ABCG2 transporters, ferrochelatase (FECH), and heme oxygenase (HO-1) as predictive biomarkers for 5-ALA-PDT. 5-amino levulinic acid 140-145 heme oxygenase 1 Homo sapiens 105-109 32731542-7 2020 The results indicate that the interaction between 147CC514 and heme-HO-1 is considerably weak, and the enzymatic activity of 147CC514 is markedly weaker than that of CPR. 147cc514 50-58 heme oxygenase 1 Homo sapiens 68-72 32223225-6 2020 Vaped GLY also causes an increase in cellular stress signals HMOX1, NQO1, and carbonylated proteins when the e-cig device is operated at high wattages. Glycerol 6-9 heme oxygenase 1 Homo sapiens 61-66 32774680-12 2020 Noteworthily, the renal Nrf2/HO-1 pathway was activated and autophagy was enhanced after hUCBMNC injection. hucbmnc 89-96 heme oxygenase 1 Homo sapiens 29-33 32774685-6 2020 Additionally, H2O2 at 1 mM significantly decreased the mRNA expression levels of Nrf2, CAT, SOD1, SOD2, HO-1, GST-pi, NQO1, and GLCM in ARPE-19 cells; however, treatment with EE-TT reversed the downregulated mRNA expression levels of all these genes induced by H2O2. Hydrogen Peroxide 14-18 heme oxygenase 1 Homo sapiens 104-108 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Heme 17-21 heme oxygenase 1 Homo sapiens 35-39 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Biliverdine 91-101 heme oxygenase 1 Homo sapiens 17-33 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Biliverdine 91-101 heme oxygenase 1 Homo sapiens 35-39 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). ferrous iron 103-115 heme oxygenase 1 Homo sapiens 17-33 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). ferrous iron 103-115 heme oxygenase 1 Homo sapiens 35-39 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 121-136 heme oxygenase 1 Homo sapiens 17-33 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 121-136 heme oxygenase 1 Homo sapiens 35-39 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 138-140 heme oxygenase 1 Homo sapiens 17-33 32717801-1 2020 Stress-inducible heme oxygenase-1 (HO-1) catalyzes the oxidative cleavage of heme yielding biliverdin, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 138-140 heme oxygenase 1 Homo sapiens 35-39 32792954-0 2020 Kaempferol Protects Blood Vessels From Damage Induced by Oxidative Stress and Inflammation in Association With the Nrf2/HO-1 Signaling Pathway. kaempferol 0-10 heme oxygenase 1 Homo sapiens 120-124 32792954-6 2020 Finally, we observed that kaempferol corrected the levels of antioxidants and elevated the protein levels of Nrf2 and HO-1 in aortic tissues and HUVECs. kaempferol 26-36 heme oxygenase 1 Homo sapiens 118-122 32792954-7 2020 Collectively, our findings prove that kaempferol protects blood vessels from damage induced by oxidative stress and inflammation and that the Nrf2/HO-1 signaling pathway plays a key role in mediating these effects. kaempferol 38-48 heme oxygenase 1 Homo sapiens 147-151 32692767-1 2020 Heme oxygenase (HO-1) mediates the enzymatic cleavage of heme, a molecule with proinflammatory and prooxidant properties. Heme 57-61 heme oxygenase 1 Homo sapiens 16-20 32530271-0 2020 Albumin Protects Lung Cells against Acrolein Cytotoxicity and Acrolein-adducted Albumin Increases Heme Oxygenase 1 Transcripts. Acrolein 62-70 heme oxygenase 1 Homo sapiens 98-114 32708430-5 2020 The inflammatory response manifested by increased cytokine levels and reactive oxygen species results in inhibition of heme oxygenase (HO-1), with a subsequent loss of cytoprotection. Oxygen 79-85 heme oxygenase 1 Homo sapiens 135-139 32530271-6 2020 In addition, albumin inhibited acrolein-induced increase of transcripts associated with cellular stress response, activating transcription factor 3 (ATF3), and antioxidant response, heme oxygenase 1 (HMOX1) in HAEC cells. Acrolein 31-39 heme oxygenase 1 Homo sapiens 182-198 32530271-6 2020 In addition, albumin inhibited acrolein-induced increase of transcripts associated with cellular stress response, activating transcription factor 3 (ATF3), and antioxidant response, heme oxygenase 1 (HMOX1) in HAEC cells. Acrolein 31-39 heme oxygenase 1 Homo sapiens 200-205 32530271-7 2020 Acrolein-adducted albumin itself increased HMOX1 transcripts but not ATF3 transcripts. Acrolein 0-8 heme oxygenase 1 Homo sapiens 43-48 32530271-8 2020 The HMOX1 transcript increase was inhibited by hydralazine, a carbonyl scavenger, suggesting that the carbonyl group of acrolein-adducted albumin mediated HMOX1 transcript increase. Hydralazine 47-58 heme oxygenase 1 Homo sapiens 4-9 32530271-8 2020 The HMOX1 transcript increase was inhibited by hydralazine, a carbonyl scavenger, suggesting that the carbonyl group of acrolein-adducted albumin mediated HMOX1 transcript increase. Hydralazine 47-58 heme oxygenase 1 Homo sapiens 155-160 32530271-8 2020 The HMOX1 transcript increase was inhibited by hydralazine, a carbonyl scavenger, suggesting that the carbonyl group of acrolein-adducted albumin mediated HMOX1 transcript increase. Acrolein 120-128 heme oxygenase 1 Homo sapiens 4-9 32530271-8 2020 The HMOX1 transcript increase was inhibited by hydralazine, a carbonyl scavenger, suggesting that the carbonyl group of acrolein-adducted albumin mediated HMOX1 transcript increase. Acrolein 120-128 heme oxygenase 1 Homo sapiens 155-160 32765809-4 2020 In this study, we firstly reported that brusatol exerted the growth-inhibitory effects on HER2-positive cancer cells by regressing Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling pathways in these cells. brusatol 40-48 heme oxygenase 1 Homo sapiens 136-140 32709140-8 2020 We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-kappaB (NF-kappaB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. N-(3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl)-2-methyl-2-((5-(trifluoromethyl)pyridin-2-yl)oxy)propanamide 41-48 heme oxygenase 1 Homo sapiens 167-183 32709140-8 2020 We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-kappaB (NF-kappaB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. N-(3-(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl)-2-methyl-2-((5-(trifluoromethyl)pyridin-2-yl)oxy)propanamide 41-48 heme oxygenase 1 Homo sapiens 185-189 32765809-0 2020 Nrf2 Inhibitor, Brusatol in Combination with Trastuzumab Exerts Synergistic Antitumor Activity in HER2-Positive Cancers by Inhibiting Nrf2/HO-1 and HER2-AKT/ERK1/2 Pathways. brusatol 16-24 heme oxygenase 1 Homo sapiens 139-143 32709140-8 2020 We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-kappaB (NF-kappaB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. ZSTK474 53-60 heme oxygenase 1 Homo sapiens 167-183 32709140-8 2020 We observed that combined treatment with AZD0364 and ZSTK474 affected nuclear factor-kappaB (NF-kappaB) and antioxidant protein levels: NF-E2-related factor 2 (NRF2), heme oxygenase-1 (HO-1), thioredoxin (Trx), thioredoxin reductase (TrxR), and the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio. ZSTK474 53-60 heme oxygenase 1 Homo sapiens 185-189 32765809-5 2020 More importantly, we found that brusatol synergistically enhanced the antitumor activity of trastuzumab against HER2-positive SK-OV-3 and BT-474 cells, which may be attributed to the inhibition of Nrf2/HO-1 and HER2-AKT/ERK1/2 signaling pathways. brusatol 32-40 heme oxygenase 1 Homo sapiens 202-206 32708634-0 2020 Cannabidiol Promotes Endothelial Cell Survival by Heme Oxygenase-1-Mediated Autophagy. Cannabidiol 0-11 heme oxygenase 1 Homo sapiens 50-66 32709035-10 2020 Conclusions: Our results suggest the presence of molecular stages of PA, defined according to the over-expression (first stage) or under-expression (second stage) of the HMOX1, SOD1, IL-6, and IFNgamma genes in abnormal skin tissue. Protactinium 69-71 heme oxygenase 1 Homo sapiens 170-175 32708634-2 2020 This study investigated the influence of CBD on the expression of heme oxygenase-1 (HO-1) and its functional role in regulating metabolic, autophagic, and apoptotic processes of human umbilical vein endothelial cells (HUVEC). Cannabidiol 41-44 heme oxygenase 1 Homo sapiens 66-82 32708634-4 2020 CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Cannabidiol 0-3 heme oxygenase 1 Homo sapiens 12-16 32708634-2 2020 This study investigated the influence of CBD on the expression of heme oxygenase-1 (HO-1) and its functional role in regulating metabolic, autophagic, and apoptotic processes of human umbilical vein endothelial cells (HUVEC). Cannabidiol 41-44 heme oxygenase 1 Homo sapiens 84-88 32708634-4 2020 CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Reactive Oxygen Species 186-189 heme oxygenase 1 Homo sapiens 12-16 32708634-3 2020 Concentrations up to 10 microM CBD showed a concentration-dependent increase of HO-1 mRNA and protein and an increase of the HO-1-regulating transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). Cannabidiol 31-34 heme oxygenase 1 Homo sapiens 80-84 32708634-4 2020 CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Acetylcysteine 201-220 heme oxygenase 1 Homo sapiens 12-16 32708634-3 2020 Concentrations up to 10 microM CBD showed a concentration-dependent increase of HO-1 mRNA and protein and an increase of the HO-1-regulating transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). Cannabidiol 31-34 heme oxygenase 1 Homo sapiens 125-129 32708634-4 2020 CBD-induced HO-1 expression was not decreased by antagonists of cannabinoid-activated receptors (CB1, CB2, transient receptor potential vanilloid 1), but by the reactive oxygen species (ROS) scavenger N-acetyl-L-cysteine (NAC). Acetylcysteine 222-225 heme oxygenase 1 Homo sapiens 12-16 32376267-0 2020 Heme oxygenase-1 alleviated non-alcoholic fatty liver disease via suppressing ROS-dependent endoplasmic reticulum stress. ros 78-81 heme oxygenase 1 Homo sapiens 0-16 32708634-9 2020 On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. tin protoporphyrin IX 56-77 heme oxygenase 1 Homo sapiens 37-41 32708634-9 2020 On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. tin protoporphyrin IX 79-85 heme oxygenase 1 Homo sapiens 37-41 32708634-9 2020 On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. Cannabidiol 167-170 heme oxygenase 1 Homo sapiens 37-41 32708634-9 2020 On the other hand, the inhibition of HO-1 activity with tin protoporphyrin IX (SnPPIX) or knockdown of HO-1 expression by Nrf2 siRNA was associated with a decrease in CBD-mediated autophagy and apoptosis. Cannabidiol 167-170 heme oxygenase 1 Homo sapiens 103-107 32708634-10 2020 In summary, our data show for the first time ROS-mediated HO-1 expression in endothelial cells as a mechanism by which CBD mediates protective autophagy, which at higher CBD concentrations, however, can no longer prevent cell death inducing apoptosis. Reactive Oxygen Species 45-48 heme oxygenase 1 Homo sapiens 58-62 32708634-10 2020 In summary, our data show for the first time ROS-mediated HO-1 expression in endothelial cells as a mechanism by which CBD mediates protective autophagy, which at higher CBD concentrations, however, can no longer prevent cell death inducing apoptosis. Cannabidiol 119-122 heme oxygenase 1 Homo sapiens 58-62 32708634-10 2020 In summary, our data show for the first time ROS-mediated HO-1 expression in endothelial cells as a mechanism by which CBD mediates protective autophagy, which at higher CBD concentrations, however, can no longer prevent cell death inducing apoptosis. Cannabidiol 170-173 heme oxygenase 1 Homo sapiens 58-62 32209424-0 2020 New insights into the protection of growth hormone in cisplatin-induced nephrotoxicity: The impact of IGF-1 on the Keap1-Nrf2/HO-1 signaling. Cisplatin 54-63 heme oxygenase 1 Homo sapiens 126-130 32209424-7 2020 Cisplatin upregulated the HMGB-1/NF-kappaB and downregulated Nrf2/HO-1 pathways which were reversed by hGH and aligned with increased renal IGF-1 expression. Cisplatin 0-9 heme oxygenase 1 Homo sapiens 66-70 32209424-10 2020 SIGNIFICANCE: these results concluded that hGH can attenuate the inflammation and oxidative stress attained by CDDP probably through inhibition of Nrf2/HO-1 pathway. Cisplatin 111-115 heme oxygenase 1 Homo sapiens 152-156 32209424-11 2020 We also suggested that Keap1/Nrf2-mediated upregulation of the antioxidant HO-1 might inhibit HMGB-1/NF-kappaB signaling and thus provide the principal protection mechanism offered by hGH against CDDP-induced kidney injury. Cisplatin 196-200 heme oxygenase 1 Homo sapiens 75-79 32335165-10 2020 Piperine inhibited iNOS expression concomitant with enhanced expression levels of Nrf2, HO1 and the total antioxidant capacity in the hippocampal tissue. piperine 0-8 heme oxygenase 1 Homo sapiens 88-91 32695819-0 2020 Antioxidant Effects and Cytoprotective Potentials of Herbal Tea against H2O2-Induced Oxidative Damage by Activating Heme Oxygenase1 Pathway. Hydrogen Peroxide 72-76 heme oxygenase 1 Homo sapiens 116-131 32760278-6 2020 The non-resolving hyper-inflammation in CF lungs is attributed to an impairment of several signaling pathways associated with resolution of the inflammatory response, including the heme oxygenase-1/carbon monoxide (HO-1/CO) pathway. Carbon Monoxide 198-213 heme oxygenase 1 Homo sapiens 181-197 32760278-6 2020 The non-resolving hyper-inflammation in CF lungs is attributed to an impairment of several signaling pathways associated with resolution of the inflammatory response, including the heme oxygenase-1/carbon monoxide (HO-1/CO) pathway. Carbon Monoxide 198-213 heme oxygenase 1 Homo sapiens 215-219 32760278-7 2020 HO-1 is an enzyme that degrades heme groups, leading to the production of potent antioxidant, anti-inflammatory, and bactericidal mediators, such as biliverdin, bilirubin, and CO. Heme 32-36 heme oxygenase 1 Homo sapiens 0-4 32760278-7 2020 HO-1 is an enzyme that degrades heme groups, leading to the production of potent antioxidant, anti-inflammatory, and bactericidal mediators, such as biliverdin, bilirubin, and CO. Biliverdine 149-159 heme oxygenase 1 Homo sapiens 0-4 32760278-7 2020 HO-1 is an enzyme that degrades heme groups, leading to the production of potent antioxidant, anti-inflammatory, and bactericidal mediators, such as biliverdin, bilirubin, and CO. Bilirubin 161-170 heme oxygenase 1 Homo sapiens 0-4 32760278-13 2020 Finally, we discuss recent studies highlighting how endogenous HO-1 can be induced by administration of controlled doses of CO to reduce lung hyper-inflammation, oxidative stress, bacterial infection, and dysfunctional ion transport, which are all hallmarks of CF lung disease. Carbon Monoxide 124-126 heme oxygenase 1 Homo sapiens 63-67 32733250-9 2020 In the tumor necrosis factor alpha (TNF)-alpha/interferon (IFN)-gamma-stimulated HaCaT cells, CBT inhibited the production of pro-inflammatory cytokines and chemokines and elevated the nuclear translocation of NF-E2 p45 related factors 2 (Nrf2) and subsequent production of heme oxygenase-1 (HO-1). N,N-BIS(4-CHLOROBENZYL)-1H-1,2,3,4-TETRAAZOL-5-AMINE 94-97 heme oxygenase 1 Homo sapiens 274-290 32733250-9 2020 In the tumor necrosis factor alpha (TNF)-alpha/interferon (IFN)-gamma-stimulated HaCaT cells, CBT inhibited the production of pro-inflammatory cytokines and chemokines and elevated the nuclear translocation of NF-E2 p45 related factors 2 (Nrf2) and subsequent production of heme oxygenase-1 (HO-1). N,N-BIS(4-CHLOROBENZYL)-1H-1,2,3,4-TETRAAZOL-5-AMINE 94-97 heme oxygenase 1 Homo sapiens 292-296 31927051-10 2020 These results indicate that nifedipine inhibited inflammation and oxidative stress in chondrocytes via activation of Nrf-2/HO-1 signaling. Nifedipine 28-38 heme oxygenase 1 Homo sapiens 123-127 32695819-6 2020 Furthermore, enrichment analysis of differentially expressed genes revealed that heme oxygenase1 (HMOX1) was a key gene for protective effect of WHT on oxidative stress-induced cell damage. wht 145-148 heme oxygenase 1 Homo sapiens 81-96 32695819-6 2020 Furthermore, enrichment analysis of differentially expressed genes revealed that heme oxygenase1 (HMOX1) was a key gene for protective effect of WHT on oxidative stress-induced cell damage. wht 145-148 heme oxygenase 1 Homo sapiens 98-103 32344355-0 2020 Akebia Saponin D ameliorated kidney injury and exerted anti-inflammatory and anti-apoptotic effects in diabetic nephropathy by activation of NRF2/HO-1 and inhibition of NF-KB pathway. akebia saponin D 0-16 heme oxygenase 1 Homo sapiens 146-150 32801467-5 2020 Furthermore, enzymatically synthesized glycogen increased in the expression level of heme oxygenase-1, NAD(P)H: quinone oxidoreductase 1, and NF-E2-related factor 2, a transcriptional factor for heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1. Glycogen 39-47 heme oxygenase 1 Homo sapiens 85-101 32319555-4 2020 CA and its natural derivative, 2-methoxycinnamaldehyde (MCA), markedly increased the cellular protein level of heme oxygenase-1 (HO-1) and promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus. 2-methoxycinnamaldehyde 31-54 heme oxygenase 1 Homo sapiens 111-127 32319555-4 2020 CA and its natural derivative, 2-methoxycinnamaldehyde (MCA), markedly increased the cellular protein level of heme oxygenase-1 (HO-1) and promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus. 2-methoxycinnamaldehyde 31-54 heme oxygenase 1 Homo sapiens 129-133 32319555-4 2020 CA and its natural derivative, 2-methoxycinnamaldehyde (MCA), markedly increased the cellular protein level of heme oxygenase-1 (HO-1) and promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus. 2-methoxycinnamaldehyde 56-59 heme oxygenase 1 Homo sapiens 111-127 32319555-4 2020 CA and its natural derivative, 2-methoxycinnamaldehyde (MCA), markedly increased the cellular protein level of heme oxygenase-1 (HO-1) and promoted the translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) to the nucleus. 2-methoxycinnamaldehyde 56-59 heme oxygenase 1 Homo sapiens 129-133 32319555-5 2020 CA-mediated Nrf2/HO-1 activation protected the HUVECs from H2O2-induced oxidative stress, which promotes apoptosis. Hydrogen Peroxide 59-63 heme oxygenase 1 Homo sapiens 17-21 32801466-10 2020 On the other hand, ESG increased nuclear translocation of Nrf2 and expression of antioxidant proteins, HO-1 and NQO1. L-Gamma-Glutamyl-S-[(3s)-1-Ethyl-2,5-Dioxopyrrolidin-3-Yl]-L-Cysteinylglycine 19-22 heme oxygenase 1 Homo sapiens 103-107 32240302-0 2020 JUND-dependent upregulation of HMOX1 is associated with cisplatin resistance in muscle-invasive bladder cancer. Cisplatin 56-65 heme oxygenase 1 Homo sapiens 31-36 32240302-6 2020 This CDDP resistance was further demonstrated to be mediated, at least in part, by transactivation of HMOX1 [the gene encoding heme oxygenase-1 (HO-1)], one of the most important antioxidant signaling pathways of cell adaptation to stress. cddp 5-9 heme oxygenase 1 Homo sapiens 102-107 32240302-6 2020 This CDDP resistance was further demonstrated to be mediated, at least in part, by transactivation of HMOX1 [the gene encoding heme oxygenase-1 (HO-1)], one of the most important antioxidant signaling pathways of cell adaptation to stress. cddp 5-9 heme oxygenase 1 Homo sapiens 127-143 32240302-6 2020 This CDDP resistance was further demonstrated to be mediated, at least in part, by transactivation of HMOX1 [the gene encoding heme oxygenase-1 (HO-1)], one of the most important antioxidant signaling pathways of cell adaptation to stress. cddp 5-9 heme oxygenase 1 Homo sapiens 145-149 32240302-7 2020 One mutation within the HMOX1 promoter successfully abolished oxidative stress-enhanced and JUND-driven HMOX1 promoter activation, suggesting that this unique site synergized for maximal HO-1 induction in CDDP-challenged BC cells. cddp 205-209 heme oxygenase 1 Homo sapiens 24-29 32240302-7 2020 One mutation within the HMOX1 promoter successfully abolished oxidative stress-enhanced and JUND-driven HMOX1 promoter activation, suggesting that this unique site synergized for maximal HO-1 induction in CDDP-challenged BC cells. cddp 205-209 heme oxygenase 1 Homo sapiens 104-109 32240302-7 2020 One mutation within the HMOX1 promoter successfully abolished oxidative stress-enhanced and JUND-driven HMOX1 promoter activation, suggesting that this unique site synergized for maximal HO-1 induction in CDDP-challenged BC cells. cddp 205-209 heme oxygenase 1 Homo sapiens 187-191 32801467-5 2020 Furthermore, enzymatically synthesized glycogen increased in the expression level of heme oxygenase-1, NAD(P)H: quinone oxidoreductase 1, and NF-E2-related factor 2, a transcriptional factor for heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1. Glycogen 39-47 heme oxygenase 1 Homo sapiens 195-211 32801467-7 2020 Knockdown of heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1 canceled enzymatically synthesized glycogen-suppressed reactive oxygen species accumulation in normal human epidermal keratinocytes. Glycogen 103-111 heme oxygenase 1 Homo sapiens 13-29 32801467-7 2020 Knockdown of heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1 canceled enzymatically synthesized glycogen-suppressed reactive oxygen species accumulation in normal human epidermal keratinocytes. Reactive Oxygen Species 123-146 heme oxygenase 1 Homo sapiens 13-29 32801467-8 2020 It is, therefore, concluded that enzymatically synthesized glycogen inhibited ultraviolet B-induced oxidative stress through increasing the expression level of heme oxygenase-1 and NAD(P)H: quinone oxidoreductase 1 through the NF-E2-related factor 2 pathway in normal human epidermal keratinocytes. Glycogen 59-67 heme oxygenase 1 Homo sapiens 160-176 32336667-7 2020 We observed a clear additive effect of O3 and DEE in combination with UV in increasing levels of several oxidative (4HNE, HO-1) and inflammatory (COX2, NF-kappaB) markers and loss of barrier-associated proteins, such as filaggrin and involucrin. Ozone 39-41 heme oxygenase 1 Homo sapiens 122-126 32319645-9 2020 Additionally, treatment with petatewalide B activated AMPK in the OGD/R-exposed SH-SY5Y cells and upregulated activation of the downstream transcription factor Nrf2, which accompanied heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) expression. petatewalide b 29-43 heme oxygenase 1 Homo sapiens 184-200 32319645-9 2020 Additionally, treatment with petatewalide B activated AMPK in the OGD/R-exposed SH-SY5Y cells and upregulated activation of the downstream transcription factor Nrf2, which accompanied heme oxygenase 1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1) expression. petatewalide b 29-43 heme oxygenase 1 Homo sapiens 202-206 32319645-10 2020 Furthermore, silencing AMPK, Nrf2, HO-1 and NQO1 expression inhibited petatewalide B"s protective effect against apoptosis in the OGD/R-exposed SH-SY5Y cells. petatewalide b 70-84 heme oxygenase 1 Homo sapiens 35-39 32629871-6 2020 NRF2 knockdown reversed 5-FU resistance of PDAC cells via suppression of ABCG2 and HO-1. Fluorouracil 24-28 heme oxygenase 1 Homo sapiens 83-87 32695110-4 2020 Heme is a chemoattractant, activates the complement system, modulates host defense mechanisms through the activation of innate immune receptors and the heme oxygenase-1/ferritin system, and induces innate immune memory. Heme 0-4 heme oxygenase 1 Homo sapiens 152-168 32655974-6 2020 However, these processes could be inactivated by compound C (inhibitor of AMPK) and zinc protoporphyrin-1 (ZnPP-1; inhibitor of HO-1). znpp-1 107-113 heme oxygenase 1 Homo sapiens 128-132 32706194-11 2020 At the same time, N-hydroxy-N"-(4-butyl-2-methylphenyl)-formamidine protects SH-SY5Y cells against oxygen-glucose deprivation and reoxygenation-induced injury partly through the inhibition of microRNA-27-a-5p and promotion of the Bach1/HO-1 signaling pathway. n-hydroxy-n"-(4-butyl-2-methylphenyl)-formamidine 18-67 heme oxygenase 1 Homo sapiens 236-240 32655823-7 2020 In addition, the activation of NF-kappaB pathway was suppressed by 4SC-202, while up-regulation of HO-1 significantly weakened the 4SC-202-induced suppression of the NF-kappaB pathway, thereby attenuating the efficacy of 4SC-202. domatinostat 131-138 heme oxygenase 1 Homo sapiens 99-103 32655974-6 2020 However, these processes could be inactivated by compound C (inhibitor of AMPK) and zinc protoporphyrin-1 (ZnPP-1; inhibitor of HO-1). zinc protoporphyrin-1 84-105 heme oxygenase 1 Homo sapiens 128-132 32630570-10 2020 After H2O2 treatment, translocation of Nrf2 into the nucleus and the subsequent increase of Heme Oxygenase-1 (HO-1) were observed earlier in CGA-treated cells. Hydrogen Peroxide 6-10 heme oxygenase 1 Homo sapiens 110-114 32581238-6 2020 Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. Hemin 21-26 heme oxygenase 1 Homo sapiens 13-17 32581238-6 2020 Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. Hemin 21-26 heme oxygenase 1 Homo sapiens 234-238 32581238-6 2020 Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. COPP protocol 30-34 heme oxygenase 1 Homo sapiens 13-17 32581238-6 2020 Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. COPP protocol 30-34 heme oxygenase 1 Homo sapiens 234-238 32581238-6 2020 Induction of HO-1 by hemin or CoPP in vitro reduced production of IFN-gamma and IL-10 from healthy human PBMCs and decreased bacterial clearance activity of whole blood from healthy controls and beta-thalassaemia, while inhibition of HO-1 by SnPP enhanced both functions in healthy controls. S-Nitroso-N-propionyl-D,L-penicillamine 242-246 heme oxygenase 1 Homo sapiens 13-17 32581238-8 2020 Our results suggest a mechanism that excess hemin of beta-thalassaemia patients is a significant cause of immune suppression via HO-1 induction and may underlie the susceptibility of these individuals to severe bacterial infection. Hemin 44-49 heme oxygenase 1 Homo sapiens 129-133 32655823-6 2020 Here, we reported that 4SC-202 could down-regulate the expression of HO-1, and up-regulation of HO-1 could significantly attenuate the 4SC-202-induced apoptosis in SKM-1 cells. domatinostat 23-30 heme oxygenase 1 Homo sapiens 69-73 32655823-6 2020 Here, we reported that 4SC-202 could down-regulate the expression of HO-1, and up-regulation of HO-1 could significantly attenuate the 4SC-202-induced apoptosis in SKM-1 cells. domatinostat 135-142 heme oxygenase 1 Homo sapiens 96-100 32655823-7 2020 In addition, the activation of NF-kappaB pathway was suppressed by 4SC-202, while up-regulation of HO-1 significantly weakened the 4SC-202-induced suppression of the NF-kappaB pathway, thereby attenuating the efficacy of 4SC-202. domatinostat 131-138 heme oxygenase 1 Homo sapiens 99-103 32595528-6 2020 Compared with OGD-treated cells, xanomeline inhibited apoptosis, reduced ROS production, attenuated the OGD-induced HIF-1alpha increase and partially reversed the reduction of HO-1, Sirtuin-1, Bcl-2, PARP, and p-Akt induced by OGD. xanomeline 33-43 heme oxygenase 1 Homo sapiens 176-180 32670766-5 2020 Herein, lipid-polymer hybrid nanoparticle (hNP) is loaded with tin mesoporphyrin (SnMP), a HO1-inhibitor, and non-covalently modified with an engineered antibody for leukemic cell-targeted delivery. lipid-polymer 8-21 heme oxygenase 1 Homo sapiens 91-94 32506967-2 2022 Although BaP is one of the most extensively studied pollutants, the underlying mechanisms through which BaP affects reactive oxygen species (ROS)/hypoxia-inducible factor 1alpha (HIF-1alpha)/heme oxygenase 1(HO-1) signaling during lung or breast carcinogenesis are not yet fully understood. Benzo(a)pyrene 104-107 heme oxygenase 1 Homo sapiens 191-207 32506967-2 2022 Although BaP is one of the most extensively studied pollutants, the underlying mechanisms through which BaP affects reactive oxygen species (ROS)/hypoxia-inducible factor 1alpha (HIF-1alpha)/heme oxygenase 1(HO-1) signaling during lung or breast carcinogenesis are not yet fully understood. Benzo(a)pyrene 104-107 heme oxygenase 1 Homo sapiens 208-212 32506967-3 2022 In this study, we analyzed the effects of 0 (control), 1, 5, or 25 microM BaP exposure on A549 and MCF-7 cancer cells, by evaluating cell viability, cell cycle, and regulatory protein expression, metabolic gene expression, and ROS/HIF-1alpha/HO-1 signaling. Benzo(a)pyrene 74-77 heme oxygenase 1 Homo sapiens 242-246 32526964-0 2020 Baicalein Inhibits Benzo[a]pyrene-Induced Toxic Response by Downregulating Src Phosphorylation and by Upregulating NRF2-HMOX1 System. baicalein 0-9 heme oxygenase 1 Homo sapiens 120-125 32526964-0 2020 Baicalein Inhibits Benzo[a]pyrene-Induced Toxic Response by Downregulating Src Phosphorylation and by Upregulating NRF2-HMOX1 System. Benzo(a)pyrene 19-33 heme oxygenase 1 Homo sapiens 120-125 32526964-6 2020 In addition, BAI activated antioxidative system nuclear factor-erythroid 2-related factor-2 (NRF2) and heme oxygenase 1 (HMOX1), leading the reduction of BaP-induced ROS production. Benzo(a)pyrene 154-157 heme oxygenase 1 Homo sapiens 103-119 32526964-6 2020 In addition, BAI activated antioxidative system nuclear factor-erythroid 2-related factor-2 (NRF2) and heme oxygenase 1 (HMOX1), leading the reduction of BaP-induced ROS production. Benzo(a)pyrene 154-157 heme oxygenase 1 Homo sapiens 121-126 32526964-6 2020 In addition, BAI activated antioxidative system nuclear factor-erythroid 2-related factor-2 (NRF2) and heme oxygenase 1 (HMOX1), leading the reduction of BaP-induced ROS production. Reactive Oxygen Species 166-169 heme oxygenase 1 Homo sapiens 121-126 32526964-9 2020 These results indicate that BAI and BAI-containing herbal drugs may be useful for inhibiting the toxic effects of BaP via dual AHR-CYP1A1-inhibiting and NRF2-HMOX1-activating activities. Benzo(a)pyrene 114-117 heme oxygenase 1 Homo sapiens 158-163 32506967-7 2022 Moreover, the induction of ROS and the modulation of HIF-1alpha and HO-1 were observed after BaP exposure. Benzo(a)pyrene 93-96 heme oxygenase 1 Homo sapiens 68-72 32506967-8 2022 Taken together, these findings suggest that BaP affects proliferation with reference to metabolic genes and ROS/HIF-1alpha/HO-1 signaling in A549 and MCF-7 cancer cells. Benzo(a)pyrene 44-47 heme oxygenase 1 Homo sapiens 123-127 32724648-4 2020 Furthermore, COS treatment significantly upregulated the expression of Nrf2 and its downstream target genes HO-1, NQO1, and CAT. carbonyl sulfide 13-16 heme oxygenase 1 Homo sapiens 108-112 32369450-3 2020 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme that catabolizes free heme. Heme 74-78 heme oxygenase 1 Homo sapiens 0-16 32369450-3 2020 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme that catabolizes free heme. Heme 74-78 heme oxygenase 1 Homo sapiens 18-22 32369450-6 2020 Herein, we show that HO-1 is highly expressed in monocytic cells in the tumor microenvironment (TME) once they differentiate into TAMs. tams 130-134 heme oxygenase 1 Homo sapiens 21-25 32670766-10 2020 Collectively, HO1-inhibiting dual cell-targeted T-hNP/SnMP has a strong potential as a novel therapeutic in AML. tin mesoporphyrin 54-58 heme oxygenase 1 Homo sapiens 14-17 32167970-15 2020 RTV-treated HIV+ women had further increases in cystatin C (n = 20; p = 0.05), with parallel elevation of HO-1. Ritonavir 0-3 heme oxygenase 1 Homo sapiens 106-110 32222619-0 2020 Chromatograpic resolution of phenylethanolic-azole racemic compounds highlighted stereoselective inhibition of heme oxygenase-1 by (R)-enantiomers. phenylethanolic 29-44 heme oxygenase 1 Homo sapiens 111-127 32222619-0 2020 Chromatograpic resolution of phenylethanolic-azole racemic compounds highlighted stereoselective inhibition of heme oxygenase-1 by (R)-enantiomers. Azoles 45-50 heme oxygenase 1 Homo sapiens 111-127 31990992-0 2020 Inhibition of microRNA-199a-5p ameliorates oxygen-glucose deprivation/reoxygenation-induced apoptosis and oxidative stress in HT22 neurons by targeting Brg1 to activate Nrf2/HO-1 signaling. Oxygen 43-49 heme oxygenase 1 Homo sapiens 174-178 32514269-0 2020 Asiaticoside inhibits TGF-beta1-induced mesothelial-mesenchymal transition and oxidative stress via the Nrf2/HO-1 signaling pathway in the human peritoneal mesothelial cell line HMrSV5. asiaticoside 0-12 heme oxygenase 1 Homo sapiens 109-113 32633385-8 2020 In addition, HBSP also increased the activity of the Nrf2/HO-1 signaling pathway and ML385 reduced the protective effect of HBSP on BEAS-2B cells. hbsp 13-17 heme oxygenase 1 Homo sapiens 58-62 32234671-0 2020 Cryptochlorogenic acid attenuates LPS-induced inflammatory response and oxidative stress via upregulation of the Nrf2/HO-1 signaling pathway in RAW 264.7 macrophages. Cryptochlorogenic acid 0-22 heme oxygenase 1 Homo sapiens 118-122 32234491-5 2020 The liver serum biomarkers such as ALT and AST, elated levels of free radicals inducing oxidative stress characterized by a surge in Nrf-2, FOXO-1 and HO-1 genes and diminution of anti-oxidant activity characterized by a decline in SOD, GPx, and CAT genes. Free Radicals 65-78 heme oxygenase 1 Homo sapiens 151-155 31820335-12 2020 Further, lutein protected high glucose-mediated down-regulation of a redox-sensitive transcription factor, Nrf2, and antioxidant enzymes, SOD2, HO-1, and catalase. Lutein 9-15 heme oxygenase 1 Homo sapiens 144-162 31820335-12 2020 Further, lutein protected high glucose-mediated down-regulation of a redox-sensitive transcription factor, Nrf2, and antioxidant enzymes, SOD2, HO-1, and catalase. Glucose 31-38 heme oxygenase 1 Homo sapiens 144-162 32108297-0 2020 The Secretion from Bone Marrow Mesenchymal Stem Cells Pretreated with Berberine Rescues Neurons with Oxidative Damage Through Activation of the Keap1-Nrf2-HO-1 Signaling Pathway. Berberine 70-79 heme oxygenase 1 Homo sapiens 155-159 32429564-6 2020 Mechanically, WFA inhibits matrix metalloproteinase (MMP)-2 and MMP-9 activities but induces mRNA expression for a group of antioxidant genes, such as nuclear factor, erythroid 2-like 2 (NFE2L2), heme oxygenase 1 (HMOX1), glutathione-disulfide reductase (GSR), and NAD(P)H quinone dehydrogenase 1 (NQO1)) in Ca9-22 cells. withaferin A 14-17 heme oxygenase 1 Homo sapiens 196-212 32546967-0 2020 Glycine Improves Ischemic Stroke Through miR-19a-3p/AMPK/GSK-3beta/HO-1 Pathway. Glycine 0-7 heme oxygenase 1 Homo sapiens 67-71 32546967-14 2020 Rescue experiments demonstrated that glycine improved cell apoptosis, inflammatory response and glucose metabolism disorder of ischemic stroke through miR-19a-3p/AMPK/GSK-3beta/HO-1 pathway. Glycine 37-44 heme oxygenase 1 Homo sapiens 177-181 32546967-15 2020 Conclusion: Glycine improves ischemic stroke through miR-19a-3p/AMPK/GSK-3beta/HO-1 pathway. Glycine 12-19 heme oxygenase 1 Homo sapiens 79-83 32536057-1 2020 Objective: To investigate the effect of rosiglitazone (RGZ) on the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and heme oxygenase-1 (HO-1) in hepatic stellate cells (HSCs). Rosiglitazone 40-53 heme oxygenase 1 Homo sapiens 164-168 32536057-1 2020 Objective: To investigate the effect of rosiglitazone (RGZ) on the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and heme oxygenase-1 (HO-1) in hepatic stellate cells (HSCs). Rosiglitazone 55-58 heme oxygenase 1 Homo sapiens 146-162 32536057-1 2020 Objective: To investigate the effect of rosiglitazone (RGZ) on the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and heme oxygenase-1 (HO-1) in hepatic stellate cells (HSCs). Rosiglitazone 55-58 heme oxygenase 1 Homo sapiens 164-168 32536057-11 2020 HO-1 mRNA (1.80 +- 0.36) and protein (0.61 +- 0.09) relative expression was significantly increased in RGZ + ZnPP-IX group as compared to blank control group (P < 0.05). Rosiglitazone 103-106 heme oxygenase 1 Homo sapiens 0-4 32536057-11 2020 HO-1 mRNA (1.80 +- 0.36) and protein (0.61 +- 0.09) relative expression was significantly increased in RGZ + ZnPP-IX group as compared to blank control group (P < 0.05). zinc protoporphyrin 109-116 heme oxygenase 1 Homo sapiens 0-4 32536057-13 2020 Conclusion: The effect of rosiglitazone on inducing increased expression of PPARgamma, and then inhibiting HSC proliferation activity and collagen production may be realized by regulating its downstream HO-1 expression. Rosiglitazone 26-39 heme oxygenase 1 Homo sapiens 203-207 32485912-0 2020 Roles of JNK/Nrf2 Pathway on Hemin-Induced Heme Oxygenase-1 Activation in MCF-7 Human Breast Cancer Cells. Hemin 29-34 heme oxygenase 1 Homo sapiens 43-59 32485912-2 2020 In this study, we investigated that the mechanisms of hemin-induced HO-1 expression and its signaling pathways in human breast cancer cell. Hemin 54-59 heme oxygenase 1 Homo sapiens 68-72 32485912-4 2020 Hemin increased HO-1 expression in MCF-7 cells in a dose- and time-dependent manner. Hemin 0-5 heme oxygenase 1 Homo sapiens 16-20 32485912-5 2020 Hemin enhanced HO-1 expression through the activation of c-Jun N-terminal kinases (JNK) signaling pathway. Hemin 0-5 heme oxygenase 1 Homo sapiens 15-19 32485912-6 2020 Hemin also induced activation of Nrf2, a major transcription factor of HO-1 expression. Hemin 0-5 heme oxygenase 1 Homo sapiens 71-75 32485912-7 2020 These responses in MCF-7 cells were completely blocked by pretreatment with brazilin, a HO-1 regulator. brazilin 76-84 heme oxygenase 1 Homo sapiens 88-92 32485912-8 2020 These results indicated that brazilin inhibits hemin-induced HO-1 expressions through inactivation of JNK/Nrf2 in MCF-7 cells. brazilin 29-37 heme oxygenase 1 Homo sapiens 61-65 32485912-8 2020 These results indicated that brazilin inhibits hemin-induced HO-1 expressions through inactivation of JNK/Nrf2 in MCF-7 cells. Hemin 47-52 heme oxygenase 1 Homo sapiens 61-65 32485912-9 2020 Thus, our findings suggest that HO-1 is an important anticancer-target of brazilin in human breast cancer. brazilin 74-82 heme oxygenase 1 Homo sapiens 32-36 32455831-4 2020 HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. Carbon Monoxide 82-97 heme oxygenase 1 Homo sapiens 0-4 32455831-4 2020 HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. Carbon Monoxide 99-101 heme oxygenase 1 Homo sapiens 0-4 32455831-4 2020 HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. ferrous iron 104-116 heme oxygenase 1 Homo sapiens 0-4 32455831-4 2020 HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. ammonium ferrous sulfate 118-122 heme oxygenase 1 Homo sapiens 0-4 32455831-4 2020 HO-1 catalyzes the enzymatic degradation of heme with the simultaneous release of carbon monoxide (CO), ferrous iron (Fe2+), and biliverdin. Biliverdine 129-139 heme oxygenase 1 Homo sapiens 0-4 32536057-0 2020 [Rosiglitazone inhibits hepatic stellate cell proliferation by regulating peroxisome proliferator-activated receptor gamma/ heme oxygenase-1 expression]. Rosiglitazone 1-14 heme oxygenase 1 Homo sapiens 124-140 32536057-1 2020 Objective: To investigate the effect of rosiglitazone (RGZ) on the expression of peroxisome proliferator-activated receptor gamma (PPARgamma) and heme oxygenase-1 (HO-1) in hepatic stellate cells (HSCs). Rosiglitazone 40-53 heme oxygenase 1 Homo sapiens 146-162 32509156-4 2020 Besides, it was also expounded that nuclear factor-erythroid 2 (NF-E2)-related factor 2/BTB domain and CNC homolog 1 (Nrf2/Bach1) might be involved in the regulation of HO-1 and the quantum effect of photon altered ROS generation. Reactive Oxygen Species 215-218 heme oxygenase 1 Homo sapiens 169-173 32429564-6 2020 Mechanically, WFA inhibits matrix metalloproteinase (MMP)-2 and MMP-9 activities but induces mRNA expression for a group of antioxidant genes, such as nuclear factor, erythroid 2-like 2 (NFE2L2), heme oxygenase 1 (HMOX1), glutathione-disulfide reductase (GSR), and NAD(P)H quinone dehydrogenase 1 (NQO1)) in Ca9-22 cells. withaferin A 14-17 heme oxygenase 1 Homo sapiens 214-219 32499710-9 2020 Comparing to the UVA radiation, PF pre-treatment could prominently increase the MTS activity, decrease cell apoptosis, reduce the generations of ROS and MDA, increase the activity of SOD and increase the expression of Nrf2 and its target genes HO-1 and NQ-O1. peoniflorin 32-34 heme oxygenase 1 Homo sapiens 244-248 32509141-0 2020 Apatinib, a Novel Tyrosine Kinase Inhibitor, Promotes ROS-Dependent Apoptosis and Autophagy via the Nrf2/HO-1 Pathway in Ovarian Cancer Cells. apatinib 0-8 heme oxygenase 1 Homo sapiens 105-109 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. apatinib 32-40 heme oxygenase 1 Homo sapiens 161-177 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. apatinib 32-40 heme oxygenase 1 Homo sapiens 179-183 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. Glutathione 52-63 heme oxygenase 1 Homo sapiens 161-177 32105681-0 2020 Upregulation of heme oxygenase-1 by Brahma-related gene 1 through Nrf2 signaling confers protective effect against high glucose-induced oxidative damage of retinal ganglion cells. Glucose 120-127 heme oxygenase 1 Homo sapiens 16-32 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. ros 76-79 heme oxygenase 1 Homo sapiens 161-177 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. ros 76-79 heme oxygenase 1 Homo sapiens 179-183 32509141-5 2020 Mechanistically, we showed that apatinib suppressed glutathione to generate ROS via the downregulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway and maintained an antitumor effect at a low level of VEGFR2 in ovarian cancer, suggesting that combination of apatinib with Nrf2 inhibitor may be a promising therapy strategy for patients with ovarian cancer. Glutathione 52-63 heme oxygenase 1 Homo sapiens 179-183 32291269-13 2020 p21 responses to high-dose SnPP and HO-1 responses to low-dose SnPP were significantly suppressed in participants with CKD versus healthy volunteers. S-Nitroso-N-propionyl-D,L-penicillamine 63-67 heme oxygenase 1 Homo sapiens 36-40 32494130-8 2020 Meanwhile, we showed that CuONPs exposure induced oxidative stress, indicated by the increase of cellular levels of superoxide anions, the upregulation of protein levels of heme oxygenase-1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), the elevation of the levels of malondialdehyde (MDA), but the reduction of glutathione to glutathione disulfide ratio. cuonps 26-32 heme oxygenase 1 Homo sapiens 173-189 32494130-8 2020 Meanwhile, we showed that CuONPs exposure induced oxidative stress, indicated by the increase of cellular levels of superoxide anions, the upregulation of protein levels of heme oxygenase-1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), the elevation of the levels of malondialdehyde (MDA), but the reduction of glutathione to glutathione disulfide ratio. cuonps 26-32 heme oxygenase 1 Homo sapiens 191-195 32393788-3 2020 The fasting-mimicking diet selectivity reverses vitamin C-induced up-regulation of heme-oxygenase-1 and ferritin in KRAS-mutant cancer cells, consequently increasing reactive iron, oxygen species, and cell death; an effect further potentiated by chemotherapy. Ascorbic Acid 48-57 heme oxygenase 1 Homo sapiens 83-99 32393788-3 2020 The fasting-mimicking diet selectivity reverses vitamin C-induced up-regulation of heme-oxygenase-1 and ferritin in KRAS-mutant cancer cells, consequently increasing reactive iron, oxygen species, and cell death; an effect further potentiated by chemotherapy. Iron 175-179 heme oxygenase 1 Homo sapiens 83-99 32397146-7 2020 Furthermore, 12-EPAHSA increased the expression of Nrf2-dependent antioxidant enzyme genes (NQO1, GCLM, GCLC, SOD-1, and HO-1). 12-EPAHSA 13-22 heme oxygenase 1 Homo sapiens 121-125 32084418-11 2020 The results showed that agomelatine and melatonin decreased neuronal injury and promoted the Nrf2-HO-1 signalling pathway. agomelatine 24-35 heme oxygenase 1 Homo sapiens 98-102 32084418-11 2020 The results showed that agomelatine and melatonin decreased neuronal injury and promoted the Nrf2-HO-1 signalling pathway. Melatonin 40-49 heme oxygenase 1 Homo sapiens 98-102 32156661-2 2020 As a response, heme induces its metabolic degradation via heme oxygenase-1 (HO-1), activated by NF-E2-related factor 2 (NRF2), the master stress response transcription factor. Heme 15-19 heme oxygenase 1 Homo sapiens 58-74 32124251-6 2020 Pathway analysis demonstrated that the ROS/Nrf2/HO-1-SOD2-NQO-1-GCLC signaling axis is a key axis through which curcumin activates the Nrf2/ARE pathway in TMJ inflammatory chondrocytes. ros 39-42 heme oxygenase 1 Homo sapiens 48-57 31900049-6 2020 Our study has shown that IMP could regulate the signaling pathways including MAPK, PI3K/Akt, NF-kappaB, and Nrf2/HO-1 to release the inflammatory responses. imperatorin 25-28 heme oxygenase 1 Homo sapiens 113-117 31900049-7 2020 IMP might attenuate airway remodeling by the down-regulation of Nrf2/HO-1/ROS/PI3K/Akt, Nrf2/HO-1/ROS/MAPK, and Nrf2/HO-1/ROS/NF-kappaB signaling pathways. imperatorin 0-3 heme oxygenase 1 Homo sapiens 69-73 31900049-7 2020 IMP might attenuate airway remodeling by the down-regulation of Nrf2/HO-1/ROS/PI3K/Akt, Nrf2/HO-1/ROS/MAPK, and Nrf2/HO-1/ROS/NF-kappaB signaling pathways. imperatorin 0-3 heme oxygenase 1 Homo sapiens 93-97 31900049-7 2020 IMP might attenuate airway remodeling by the down-regulation of Nrf2/HO-1/ROS/PI3K/Akt, Nrf2/HO-1/ROS/MAPK, and Nrf2/HO-1/ROS/NF-kappaB signaling pathways. imperatorin 0-3 heme oxygenase 1 Homo sapiens 93-97 32124251-6 2020 Pathway analysis demonstrated that the ROS/Nrf2/HO-1-SOD2-NQO-1-GCLC signaling axis is a key axis through which curcumin activates the Nrf2/ARE pathway in TMJ inflammatory chondrocytes. Curcumin 112-120 heme oxygenase 1 Homo sapiens 48-57 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Heme 44-48 heme oxygenase 1 Homo sapiens 11-27 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Heme 44-48 heme oxygenase 1 Homo sapiens 29-33 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Carbon Monoxide 52-54 heme oxygenase 1 Homo sapiens 11-27 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Carbon Monoxide 52-54 heme oxygenase 1 Homo sapiens 29-33 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Iron 56-60 heme oxygenase 1 Homo sapiens 11-27 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Iron 56-60 heme oxygenase 1 Homo sapiens 29-33 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Biliverdine 66-76 heme oxygenase 1 Homo sapiens 11-27 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Biliverdine 66-76 heme oxygenase 1 Homo sapiens 29-33 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Bilirubin 77-86 heme oxygenase 1 Homo sapiens 11-27 32006543-1 2020 OBJECTIVE: Heme oxygenase-1 (HO-1) degrades heme to CO, iron, and biliverdin/bilirubin. Bilirubin 77-86 heme oxygenase 1 Homo sapiens 29-33 32006543-2 2020 Although serum bilirubin levels were often reported in patients with coronary artery disease (CAD), HO-1 levels in patients with CAD and the association between HO-1 and bilirubin levels have not been clarified. Bilirubin 170-179 heme oxygenase 1 Homo sapiens 161-165 32006543-8 2020 In multivariate analysis, HO-1 levels were a significant factor for CAD independent of atherosclerotic risk factors and bilirulin levels. bilirulin 120-129 heme oxygenase 1 Homo sapiens 26-30 32112801-4 2020 This was associated with reduced ferritin induction by hemoglobin; expression of heme oxygenase-1, which catalyzes iron release from heme, was not altered. Iron 115-119 heme oxygenase 1 Homo sapiens 81-97 32004978-8 2020 We observed that brusatol, an NRF2 inhibitor, as well as small interfering RNA (siRNA)-mediated knockdown of NRF2 in DLD-1 cells suppressed compound 3-induced HO-1 expression. brusatol 17-25 heme oxygenase 1 Homo sapiens 159-163 32363357-7 2020 Quercetin added apically to the endothelial cells upregulated HO-1 and downregulated PDK4 both in monoculture and in co-culture, but the total PDK4 levels were higher in the presence of HepG2 cells. Quercetin 0-9 heme oxygenase 1 Homo sapiens 62-66 32278282-0 2020 Canagliflozin inhibits vascular smooth muscle cell proliferation and migration: Role of heme oxygenase-1. Canagliflozin 0-13 heme oxygenase 1 Homo sapiens 88-104 32108290-8 2020 Our results showed that hemin was able to induce both HO-1 gene and protein expression in a cell-dependent manner being A172 more responsive to pharmacological upregulation of HO-1. Hemin 24-29 heme oxygenase 1 Homo sapiens 54-58 32108290-8 2020 Our results showed that hemin was able to induce both HO-1 gene and protein expression in a cell-dependent manner being A172 more responsive to pharmacological upregulation of HO-1. Hemin 24-29 heme oxygenase 1 Homo sapiens 176-180 32278282-5 2020 Canagliflozin also resulted in the induction of heme oxygenase-1 (HO-1) expression, and a rise in HO activity in vascular SMCs, whereas, empagliflozin or dapagliflozin had no effect on HO activity. Canagliflozin 0-13 heme oxygenase 1 Homo sapiens 48-64 32278282-5 2020 Canagliflozin also resulted in the induction of heme oxygenase-1 (HO-1) expression, and a rise in HO activity in vascular SMCs, whereas, empagliflozin or dapagliflozin had no effect on HO activity. Canagliflozin 0-13 heme oxygenase 1 Homo sapiens 66-70 32278282-6 2020 Canagliflozin also activated the HO-1 promoter and this was abrogated by mutating the antioxidant responsive element or by overexpressing dominant-negative NF-E2-related factor-2 (Nrf2). Canagliflozin 0-13 heme oxygenase 1 Homo sapiens 33-37 32278282-7 2020 The induction of HO-1 by canagliflozin relied on reactive oxygen species (ROS) formation and was negated by antioxidants. Canagliflozin 25-38 heme oxygenase 1 Homo sapiens 17-21 32278282-7 2020 The induction of HO-1 by canagliflozin relied on reactive oxygen species (ROS) formation and was negated by antioxidants. Reactive Oxygen Species 49-72 heme oxygenase 1 Homo sapiens 17-21 32278282-7 2020 The induction of HO-1 by canagliflozin relied on reactive oxygen species (ROS) formation and was negated by antioxidants. Reactive Oxygen Species 74-77 heme oxygenase 1 Homo sapiens 17-21 32278282-8 2020 Finally, silencing HO-1 expression partially rescued the proliferative and migratory response of canagliflozin-treated SMCs, and this was reversed by carbon monoxide and bilirubin. Canagliflozin 97-110 heme oxygenase 1 Homo sapiens 19-23 32278282-8 2020 Finally, silencing HO-1 expression partially rescued the proliferative and migratory response of canagliflozin-treated SMCs, and this was reversed by carbon monoxide and bilirubin. Carbon Monoxide 150-165 heme oxygenase 1 Homo sapiens 19-23 32278282-8 2020 Finally, silencing HO-1 expression partially rescued the proliferative and migratory response of canagliflozin-treated SMCs, and this was reversed by carbon monoxide and bilirubin. Bilirubin 170-179 heme oxygenase 1 Homo sapiens 19-23 32278282-10 2020 Moreover, it demonstrates that canagliflozin stimulates the expression of HO-1 in vascular SMCs via the ROS-Nrf2 pathway, and that the induction of HO-1 contributes to the cellular actions of canagliflozin. Canagliflozin 31-44 heme oxygenase 1 Homo sapiens 74-78 32278282-10 2020 Moreover, it demonstrates that canagliflozin stimulates the expression of HO-1 in vascular SMCs via the ROS-Nrf2 pathway, and that the induction of HO-1 contributes to the cellular actions of canagliflozin. Reactive Oxygen Species 104-107 heme oxygenase 1 Homo sapiens 74-78 32278282-10 2020 Moreover, it demonstrates that canagliflozin stimulates the expression of HO-1 in vascular SMCs via the ROS-Nrf2 pathway, and that the induction of HO-1 contributes to the cellular actions of canagliflozin. Canagliflozin 192-205 heme oxygenase 1 Homo sapiens 148-152 32411596-9 2020 Importantly, SOX8 might be a potential regulator of reactive oxygen species (ROS) homeostasis, as SOX8 regulated the expression of antioxidant enzymes (GPX1, HMOX1) and reduced drug-induced ROS accumulation in GTN cell models. Reactive Oxygen Species 52-75 heme oxygenase 1 Homo sapiens 158-163 32410998-0 2020 Paeoniflorin Resists H2O2-Induced Oxidative Stress in Melanocytes by JNK/Nrf2/HO-1 Pathway. peoniflorin 0-12 heme oxygenase 1 Homo sapiens 78-82 32410998-0 2020 Paeoniflorin Resists H2O2-Induced Oxidative Stress in Melanocytes by JNK/Nrf2/HO-1 Pathway. Hydrogen Peroxide 21-25 heme oxygenase 1 Homo sapiens 78-82 32410998-5 2020 Furthermore, PF activated c-Jun amino terminal kinase (JNK) and the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway to counteract H2O2-induced oxidative damage in PIG1 and PIG3V. Hydrogen Peroxide 156-160 heme oxygenase 1 Homo sapiens 110-126 32410998-5 2020 Furthermore, PF activated c-Jun amino terminal kinase (JNK) and the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway to counteract H2O2-induced oxidative damage in PIG1 and PIG3V. Hydrogen Peroxide 156-160 heme oxygenase 1 Homo sapiens 128-132 32410998-6 2020 Taken together, our study firstly demonstrates that PF resists H2O2-induced oxidative stress in melanocytes probably by activating JNK/Nrf2/HO-1 signaling, suggesting a potential therapeutic application of PF on vitiligo. Hydrogen Peroxide 63-67 heme oxygenase 1 Homo sapiens 140-144 32411596-9 2020 Importantly, SOX8 might be a potential regulator of reactive oxygen species (ROS) homeostasis, as SOX8 regulated the expression of antioxidant enzymes (GPX1, HMOX1) and reduced drug-induced ROS accumulation in GTN cell models. Reactive Oxygen Species 77-80 heme oxygenase 1 Homo sapiens 158-163 32411596-9 2020 Importantly, SOX8 might be a potential regulator of reactive oxygen species (ROS) homeostasis, as SOX8 regulated the expression of antioxidant enzymes (GPX1, HMOX1) and reduced drug-induced ROS accumulation in GTN cell models. Reactive Oxygen Species 190-193 heme oxygenase 1 Homo sapiens 158-163 32298237-5 2020 Downregulation of HO-1 using pharmacological inhibitor ZnPPIX or siRNA knockdown significantly enhanced myeloma cell sensitivity to bortezomib in human primary CD138+ cells, U266 and RPMI8226 cell lines. Bortezomib 132-142 heme oxygenase 1 Homo sapiens 18-22 32361557-16 2020 4,5-CQME also regulated the Keap1/Nrf2 signaling pathway and enhanced both the mRNA and protein expressions of HO-1 and NQO1. 4,5-cqme 0-8 heme oxygenase 1 Homo sapiens 111-115 32387991-7 2020 Moreover, the nuclear factor E2 related factor (Nrf2) protein expression in the ferulic acid group positively correlated with the HO-1, GCLC and NQO1 protein levels. ferulic acid 80-92 heme oxygenase 1 Homo sapiens 130-134 32298237-0 2020 Heme oxygenase-1 inhibition mediates Gas6 to enhance bortezomib-sensitivity in multiple myeloma via ERK/STAT3 axis. Bortezomib 53-63 heme oxygenase 1 Homo sapiens 0-16 32298237-3 2020 Here we aim to investigate the role and mechanism of HO-1 in bortezomib-sensitivity to myeloma cells. Bortezomib 61-71 heme oxygenase 1 Homo sapiens 53-57 32410860-0 2020 Carbon Monoxide-Releasing Molecule-3 Suppresses Tumor Necrosis Factor-alpha- and Interleukin-1beta-Induced Expression of Junctional Molecules on Human Gingival Fibroblasts via the Heme Oxygenase-1 Pathway. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 180-196 32321531-0 2020 Alantolactone suppresses inflammation, apoptosis and oxidative stress in cigarette smoke-induced human bronchial epithelial cells through activation of Nrf2/HO-1 and inhibition of the NF-kappaB pathways. alantolactone 0-13 heme oxygenase 1 Homo sapiens 157-161 32298237-7 2020 Combination with HO-1 inhibition increased bortezomib-induced apoptosis and antiproliferative effects via suppressing Gas6 production. Bortezomib 43-53 heme oxygenase 1 Homo sapiens 17-21 32298237-5 2020 Downregulation of HO-1 using pharmacological inhibitor ZnPPIX or siRNA knockdown significantly enhanced myeloma cell sensitivity to bortezomib in human primary CD138+ cells, U266 and RPMI8226 cell lines. zinc protoporphyrin 55-61 heme oxygenase 1 Homo sapiens 18-22 32298237-8 2020 These findings suggest that combination of bortezomib and HO-1 inhibitor may serve as a promising therapeutic target against bortezomib-resistant MM. Bortezomib 125-135 heme oxygenase 1 Homo sapiens 58-62 32308771-10 2020 In vitro, beta-elemene in combination with cetuximab was shown to induce iron-dependent reactive oxygen species (ROS) accumulation, glutathione (GSH) depletion, lipid peroxidation, upregulation of HO-1 and transferrin, and downregulation of negative regulatory proteins for ferroptosis (GPX4, SLC7A11, FTH1, glutaminase, and SLC40A1) in KRAS mutant CRC cells. beta-elemene 10-22 heme oxygenase 1 Homo sapiens 197-201 32264868-8 2020 EMPA and DAPA (100 muM) significantly reduced SA-induced inflammation (IL1beta, TNFalpha, MCP1), oxidant stress (SOD2, TXN, HO1), but not apoptosis in MAC. empagliflozin 0-4 heme oxygenase 1 Homo sapiens 124-127 32264868-8 2020 EMPA and DAPA (100 muM) significantly reduced SA-induced inflammation (IL1beta, TNFalpha, MCP1), oxidant stress (SOD2, TXN, HO1), but not apoptosis in MAC. dapagliflozin 9-13 heme oxygenase 1 Homo sapiens 124-127 32293500-7 2020 BIX-01294 treatment or siRNA-mediated G9a knockdown led to the activation of the PERK/NRF2 pathway and HO-1 upregulation in KG1 cells. BIX 01294 0-9 heme oxygenase 1 Homo sapiens 103-107 32377300-5 2020 According to recently published data, increased endogenous CO production by inducible HO-1, its delivery by novel pharmacological CO-releasing agents, or even the direct inhalation of CO has been considered a promising alternative in future experimental and clinical therapies against various GI disorders. Carbon Monoxide 59-61 heme oxygenase 1 Homo sapiens 86-90 32334435-11 2020 Resveratrol in combination with HQ significantly upregulated HO-1 mRNA expression above that of HQ-treated cells alone. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 61-65 31981945-11 2020 In particular, OXC treatment significantly enhanced expressions of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream protein heme oxygenase-1 in ischemic CA1. Oxcarbazepine 15-18 heme oxygenase 1 Homo sapiens 145-161 32228565-0 2020 Activation of the Nrf2/HO-1 pathway by curcumin inhibits oxidative stress in human nasal fibroblasts exposed to urban particulate matter. Curcumin 39-47 heme oxygenase 1 Homo sapiens 23-27 31964465-0 2020 15d-PGJ2 inhibits NF-kappaB and AP-1-mediated MMP-9 expression and invasion of breast cancer cell by means of a heme oxygenase-1-dependent mechanism. 15-deoxyprostaglandin J2 0-8 heme oxygenase 1 Homo sapiens 112-128 31964465-5 2020 In this study, we investigated the effects of induction of HO-1 by PPARgamma on TPAinduced MMP-9 expression and cell invasion using MCF-7 breast cancer cells. tpainduced 80-90 heme oxygenase 1 Homo sapiens 59-63 31964465-8 2020 15d-PGJ2 induced HO-1 expression in a dose-dependent manner. 15-deoxyprostaglandin J2 0-8 heme oxygenase 1 Homo sapiens 17-21 31964465-9 2020 Interestingly, HO-1 siRNA significantly attenuated the inhibition of TPA-induced MMP-9 protein expression and cell invasion by 15d-PGJ2. 15-deoxyprostaglandin J2 127-135 heme oxygenase 1 Homo sapiens 15-19 31964465-10 2020 These results suggest that 15d-PGJ2 inhibits TPA-induced MMP- 9 expression and invasion of MCF-7 cells by means of a heme oxygenase-1-dependent mechanism. 15-deoxyprostaglandin J2 27-35 heme oxygenase 1 Homo sapiens 117-133 31978617-9 2020 In addition, RSV reduced the levels of senescence-associated secretory phenotype (SASP), gene markers associated with senescence (P53, P16, and P21), intracellular ROS levels and increased gene expression of enzymes protecting cells from oxidative damage (HMOX1 and SOD3). resveratrol 13-16 heme oxygenase 1 Homo sapiens 256-261 32023790-10 2020 In addition, BPA treatment enhanced gene expression of antioxidant enzymes, heme oxygenase(HO)-1, catalase, superoxide dismutase(SOD) 1 and 2. bisphenol A 13-16 heme oxygenase 1 Homo sapiens 76-106 31376303-0 2020 HO-1 downregulation favors BRAFV600 melanoma cell death induced by Vemurafenib/PLX4032 and increases NK recognition. Vemurafenib 67-78 heme oxygenase 1 Homo sapiens 0-4 31376303-0 2020 HO-1 downregulation favors BRAFV600 melanoma cell death induced by Vemurafenib/PLX4032 and increases NK recognition. Vemurafenib 79-86 heme oxygenase 1 Homo sapiens 0-4 31376303-4 2020 We have investigated the role of HO-1 expression in Vemurafenib-treated BRAFV600 melanoma cells in modulating their susceptibility to NK cell-mediated recognition. Vemurafenib 52-63 heme oxygenase 1 Homo sapiens 33-37 31376303-7 2020 HO-1 silencing/inhibition was able to induce a further significant reduction of Vemurafenib-treated melanoma viability. Vemurafenib 80-91 heme oxygenase 1 Homo sapiens 0-4 31376303-9 2020 Furthermore, melanoma cell treatment with Vemurafenib downregulated the expression of ligands of NKp30 and NKG2D activating receptors, and HO-1 silencing/inhibition was able to restore their expression. Vemurafenib 42-53 heme oxygenase 1 Homo sapiens 107-143 31376303-10 2020 Our results indicate that HO-1 downregulation can both improve the efficacy of Vemurafenib on melanoma cells and favor melanoma susceptibility to NK cell-mediated recognition and killing. Vemurafenib 79-90 heme oxygenase 1 Homo sapiens 26-30 31895874-9 2020 In addition, EGCG prevents enhanced oxidative stress via the Nrf2/HO-1 pathway. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 66-70 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 21-25 heme oxygenase 1 Homo sapiens 154-170 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 21-25 heme oxygenase 1 Homo sapiens 172-176 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 154-170 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 172-176 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 154-170 32163836-7 2020 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase (FECH) and heme oxygenase-1 (HO-1) and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 172-176 32163836-8 2020 Based on the availability of compounds targeting these proteins and inhibiting them, in this study we used modulators such as genistein, an isoflavone able to inhibit ABCG2; deferoxamine, which chelate iron ions impairing FECH activity and tin protoporphyrin IX (SnPP), the specific HO-1 inhibitor. Genistein 126-135 heme oxygenase 1 Homo sapiens 283-287 32163836-8 2020 Based on the availability of compounds targeting these proteins and inhibiting them, in this study we used modulators such as genistein, an isoflavone able to inhibit ABCG2; deferoxamine, which chelate iron ions impairing FECH activity and tin protoporphyrin IX (SnPP), the specific HO-1 inhibitor. Deferoxamine 174-186 heme oxygenase 1 Homo sapiens 283-287 32420127-0 2020 Proteomic analysis of oxidative stress response in human umbilical vein endothelial cells (HUVECs): role of heme oxygenase 1 (HMOX1) in hypoxanthine-induced oxidative stress in HUVECs. Hypoxanthine 136-148 heme oxygenase 1 Homo sapiens 108-124 32420127-0 2020 Proteomic analysis of oxidative stress response in human umbilical vein endothelial cells (HUVECs): role of heme oxygenase 1 (HMOX1) in hypoxanthine-induced oxidative stress in HUVECs. Hypoxanthine 136-148 heme oxygenase 1 Homo sapiens 126-131 32420127-11 2020 Conclusions: We found that HMOX1 was closely related to the oxidative stress response induced by hypoxanthine. Hypoxanthine 97-109 heme oxygenase 1 Homo sapiens 27-32 31868938-7 2020 Furthermore, GCN2 knockdown reduced cell apoptosis and enhanced the activation of nuclear factor E2-related factor 2/heme oxygenase-1 (Nrf2/HO-1) pathway in high glucose-stimulated ARPE-19 cells. Glucose 162-169 heme oxygenase 1 Homo sapiens 140-144 31868938-9 2020 Taken together, our findings suggest that knockdown of GCN2 inhibits high glucose-induced oxidative stress and apoptosis in ARPE-19 cells through activation of the Nrf2/HO-1 pathway. Glucose 74-81 heme oxygenase 1 Homo sapiens 169-173 31541463-0 2020 Acetaminophen sensitizing erastin-induced ferroptosis via modulation of Nrf2/heme oxygenase-1 signaling pathway in non-small-cell lung cancer. Acetaminophen 0-13 heme oxygenase 1 Homo sapiens 77-93 31541463-6 2020 As a downstream gene of Nrf2, heme oxygenase-1 expression decreased significantly with the cotreatment of erastin and APAP, which could be rescued by BM. Acetaminophen 118-122 heme oxygenase 1 Homo sapiens 30-46 31541463-6 2020 As a downstream gene of Nrf2, heme oxygenase-1 expression decreased significantly with the cotreatment of erastin and APAP, which could be rescued by BM. bardoxolone methyl 150-152 heme oxygenase 1 Homo sapiens 30-46 32259676-12 2020 Moreover, limonin induced activation of Nrf2 and increased protein expression and mRNA levels of its downstream targets, including HO-1, NQO1 and GCLC/GCLM. limonin 10-17 heme oxygenase 1 Homo sapiens 131-135 32228565-10 2020 Nrf2 production was also promoted to increase the expression of HO-1 and SOD2 by curcumin. Curcumin 81-89 heme oxygenase 1 Homo sapiens 64-68 32225089-7 2020 Oxidative stress response was also impaired in HACE1 fibroblasts, as shown by the reduced NQO1 and Hmox1 mRNA levels observed in H2O2-treated cells. Hydrogen Peroxide 129-133 heme oxygenase 1 Homo sapiens 99-104 32231654-5 2020 The results demonstrated that induction of HO-1 via cobalt-protoporphyrin (CoPP) markedly suppressed PRV replication, while HO-1 specific small interfering RNA or inhibitor zinc-protoporphyrin partially reversed the inhibitory effect of CoPP on PRV replication. cobaltiprotoporphyrin 52-73 heme oxygenase 1 Homo sapiens 43-47 32204510-4 2020 Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. Reactive Oxygen Species 113-136 heme oxygenase 1 Homo sapiens 26-30 32204510-4 2020 Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. Reactive Oxygen Species 138-141 heme oxygenase 1 Homo sapiens 26-30 32204510-4 2020 Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. Hydrogen Peroxide 154-158 heme oxygenase 1 Homo sapiens 26-30 32204510-4 2020 Ectopic overexpression of HO-1 not only attenuated cell proliferation in vitro and in vivo, but also blocked the reactive oxygen species (ROS) induced by H2O2 and the pyocyanin in HepG2 or Hep3B cells. Pyocyanine 167-176 heme oxygenase 1 Homo sapiens 26-30 32188144-4 2020 Mechanistic investigations found that the downregulation of cellular IAP 1 (cIAP1)/X-chromosome-linked IAP (XIAP) and upregulation of heme oxygenase-1 (HO-1) were critical for DMC-induced caspase-8/-9/-3 activation and apoptotic cell death. demethoxycurcumin 176-179 heme oxygenase 1 Homo sapiens 134-150 32188144-4 2020 Mechanistic investigations found that the downregulation of cellular IAP 1 (cIAP1)/X-chromosome-linked IAP (XIAP) and upregulation of heme oxygenase-1 (HO-1) were critical for DMC-induced caspase-8/-9/-3 activation and apoptotic cell death. demethoxycurcumin 176-179 heme oxygenase 1 Homo sapiens 152-156 32188144-5 2020 Moreover, p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase (JNK)1/2 were activated by DMC treatment in OSCC cells, and only the inhibition of p38 MAPK significantly abolished DMC-induced HO-1 expression and caspase-8/-9/-3 activation. demethoxycurcumin 109-112 heme oxygenase 1 Homo sapiens 210-214 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Heme 52-56 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Heme 52-56 heme oxygenase 1 Homo sapiens 18-22 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Biliverdine 62-72 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Biliverdine 62-72 heme oxygenase 1 Homo sapiens 18-22 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Biliverdine 74-76 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Biliverdine 74-76 heme oxygenase 1 Homo sapiens 18-22 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Iron 79-83 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Iron 79-83 heme oxygenase 1 Homo sapiens 18-22 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 18-22 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Carbon Monoxide 105-107 heme oxygenase 1 Homo sapiens 0-16 32231654-2 2020 Heme oxygenase-1 (HO-1) catalyzes the conversion of heme into biliverdin (BV), iron and carbon monoxide (CO), all of which have been demonstrated to protect cells from various stressors. Carbon Monoxide 105-107 heme oxygenase 1 Homo sapiens 18-22 32231654-5 2020 The results demonstrated that induction of HO-1 via cobalt-protoporphyrin (CoPP) markedly suppressed PRV replication, while HO-1 specific small interfering RNA or inhibitor zinc-protoporphyrin partially reversed the inhibitory effect of CoPP on PRV replication. cobaltiprotoporphyrin 75-79 heme oxygenase 1 Homo sapiens 43-47 32168943-0 2020 New Arylethanolimidazole Derivatives as HO-1 Inhibitors with Cytotoxicity against MCF-7 Breast Cancer Cells. arylethanolimidazole 4-24 heme oxygenase 1 Homo sapiens 40-44 32168943-1 2020 In this paper, a novel series of imidazole-based heme oxygenase-1 (HO-1) inhibitors is reported. imidazole 33-42 heme oxygenase 1 Homo sapiens 49-65 32168943-1 2020 In this paper, a novel series of imidazole-based heme oxygenase-1 (HO-1) inhibitors is reported. imidazole 33-42 heme oxygenase 1 Homo sapiens 67-71 31604020-0 2020 Salvianolic acid B exerts a protective effect in acute liver injury by regulating the Nrf2/HO-1 signaling pathway. salvianolic acid B 0-18 heme oxygenase 1 Homo sapiens 91-95 31972171-2 2020 Treatment of mouse epidermal JB-6 cells with curcumin resulted in the induction of HO-1 expression, and this was abrogated in cells transiently transfected with Nrf2 siRNA. Curcumin 45-53 heme oxygenase 1 Homo sapiens 83-87 32121588-1 2020 Mevastatin (MVS) has been previously shown to induce heme oxygenase (HO)-1 expression through Nox/ROS-dependent PDGFRalpha/PI3K/Akt/Nrf2/ARE axis in human pulmonary alveolar epithelial cells (HPAEpiCs). mevastatin 0-10 heme oxygenase 1 Homo sapiens 53-74 32121588-1 2020 Mevastatin (MVS) has been previously shown to induce heme oxygenase (HO)-1 expression through Nox/ROS-dependent PDGFRalpha/PI3K/Akt/Nrf2/ARE axis in human pulmonary alveolar epithelial cells (HPAEpiCs). mevastatin 12-15 heme oxygenase 1 Homo sapiens 53-74 32121588-1 2020 Mevastatin (MVS) has been previously shown to induce heme oxygenase (HO)-1 expression through Nox/ROS-dependent PDGFRalpha/PI3K/Akt/Nrf2/ARE axis in human pulmonary alveolar epithelial cells (HPAEpiCs). ros 98-101 heme oxygenase 1 Homo sapiens 53-74 31846839-10 2020 Vinp reduced activation of NF-kappaB, MAPK, and AKT signaling during osteoclastogenesis and prevented the production of reactive oxygen species with increased nuclear factor erythroid 2-related factor 2, heme oxygenase 1, and NAD(P)H:quinone acceptor oxidoreductase 1 expression. vinpocetine 0-4 heme oxygenase 1 Homo sapiens 204-220 31604020-3 2020 The objective of this study was to investigate the underlying mechanism of Sal B in CCl4-induced acute liver injury, especially its effect on the Nrf2/HO-1 signaling pathway. salvianolic acid B 75-80 heme oxygenase 1 Homo sapiens 151-155 31981961-8 2020 The miR-548a-3p might regulate the cellular response to hydrogen peroxide through HO-1 and HIF-1pathway. Hydrogen Peroxide 56-73 heme oxygenase 1 Homo sapiens 82-86 32111854-5 2020 The autophagy inhibitor bafilomycin caused a significant decrease in the production of Nrf2, HO-1 and NQO1 compared to DEPs treatment, whereas the Nrf2 activator sulforaphane increased the LC3B (p = 0.020) levels. bafilomycin 24-35 heme oxygenase 1 Homo sapiens 93-97 32056044-0 2020 Non-competitive heme oxygenase-1 activity inhibitor reduces non-small cell lung cancer glutathione content and regulates cell proliferation. Glutathione 87-98 heme oxygenase 1 Homo sapiens 16-32 32056044-4 2020 NSCLC cell line (A549) was treated with the HO-1 activity inhibitor VP13/47 (10 microM) and we further evaluated cell viability, apoptosis, mitochondrial dysfunction and oxidative stress. vp13/47 68-75 heme oxygenase 1 Homo sapiens 44-48 31965713-7 2020 In addition, TMS could activate ERK/Nrf2/HO-1 signaling which increased the expression of ATP-binding cassette transporters. Adenosine Triphosphate 90-93 heme oxygenase 1 Homo sapiens 41-45 31972252-6 2020 In high glucose-incubated vascular endothelial cells, NO donor attenuated oxidative stress and endothelial dysfunction via the inhibition of NADPH oxidase, where a heme oxygenase-1 (HO-1)-dependent mechanism was demonstrated for the antioxidant abilities of NO. Glucose 8-15 heme oxygenase 1 Homo sapiens 164-180 31972252-6 2020 In high glucose-incubated vascular endothelial cells, NO donor attenuated oxidative stress and endothelial dysfunction via the inhibition of NADPH oxidase, where a heme oxygenase-1 (HO-1)-dependent mechanism was demonstrated for the antioxidant abilities of NO. NADP 141-146 heme oxygenase 1 Homo sapiens 164-180 32274105-8 2020 Also, Sch A downregulated the expression of IL-8 and upregulated the expression of HO-1 mRNA in lung epithelial cells and cell supernatants, and resulted in the downregulation of the protein expression level of phosphorylated nuclear factor-kappaB. schizandrin A 6-11 heme oxygenase 1 Homo sapiens 83-87 32035862-2 2020 In living organisms, it is produced endogenously during the degradation of heme by oxygenase, which occurs in three isoforms: HO-1, HO-2 and HO-3. Heme 75-79 heme oxygenase 1 Homo sapiens 126-130 32106617-3 2020 Pretreatment with blueberry anthocyanin extract, malvidin, malvidin-3-glucoside, and malvidin-3-galactoside significantly ameliorated high-glucose-induced damage by enhancing endogenous antioxidant superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), lowering reactive oxygen species (ROS) generation and NADPH oxidase isoform 4 (NOX4) expression, and increasing the cell vitalities. Anthocyanins 28-39 heme oxygenase 1 Homo sapiens 229-245 32106617-3 2020 Pretreatment with blueberry anthocyanin extract, malvidin, malvidin-3-glucoside, and malvidin-3-galactoside significantly ameliorated high-glucose-induced damage by enhancing endogenous antioxidant superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), lowering reactive oxygen species (ROS) generation and NADPH oxidase isoform 4 (NOX4) expression, and increasing the cell vitalities. Glucose 139-146 heme oxygenase 1 Homo sapiens 229-245 32318125-0 2020 Erratum to "Tetrandrine Ameliorates Airway Remodeling of Chronic Asthma by Interfering TGF-beta1/Nrf-2/HO-1 Signaling Pathway-Mediated Oxidative Stress". tetrandrine 12-23 heme oxygenase 1 Homo sapiens 103-107 32057253-6 2020 Astragaloside IV could increase the expression of HO-1 and Nrf2 for promoting the oxidant protective effect.Conclusion: Aastragaloside IV has an anti-inflammatory and oxidative effect via regulated NF-kappaB, MAPK and HO-1/Nrf2 signalling pathways in human bronchial epithelial cells. aastragaloside 120-134 heme oxygenase 1 Homo sapiens 218-222 32057253-6 2020 Astragaloside IV could increase the expression of HO-1 and Nrf2 for promoting the oxidant protective effect.Conclusion: Aastragaloside IV has an anti-inflammatory and oxidative effect via regulated NF-kappaB, MAPK and HO-1/Nrf2 signalling pathways in human bronchial epithelial cells. astragaloside A 0-13 heme oxygenase 1 Homo sapiens 50-54 32057253-6 2020 Astragaloside IV could increase the expression of HO-1 and Nrf2 for promoting the oxidant protective effect.Conclusion: Aastragaloside IV has an anti-inflammatory and oxidative effect via regulated NF-kappaB, MAPK and HO-1/Nrf2 signalling pathways in human bronchial epithelial cells. astragaloside A 0-13 heme oxygenase 1 Homo sapiens 218-222 32015215-5 2020 These effects were reversed by treatment with the early autophagy inhibitor 3-methyl adenine, which suggests HO-1-induced autophagy suppresses IL-1beta-induced apoptosis in NPCs. 3-methyladenine 76-92 heme oxygenase 1 Homo sapiens 109-113 32057253-6 2020 Astragaloside IV could increase the expression of HO-1 and Nrf2 for promoting the oxidant protective effect.Conclusion: Aastragaloside IV has an anti-inflammatory and oxidative effect via regulated NF-kappaB, MAPK and HO-1/Nrf2 signalling pathways in human bronchial epithelial cells. aastragaloside 120-134 heme oxygenase 1 Homo sapiens 50-54 32075047-7 2020 RosA promotes the expression of Heme oxygenase-1 (HO-1) and reduces the production of reactive oxygen species (Ros), which is induced by Dox. rosmarinic acid 0-4 heme oxygenase 1 Homo sapiens 32-48 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Heme 83-87 heme oxygenase 1 Homo sapiens 0-16 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Heme 83-87 heme oxygenase 1 Homo sapiens 18-22 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 0-16 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 18-22 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Iron 110-114 heme oxygenase 1 Homo sapiens 0-16 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Iron 110-114 heme oxygenase 1 Homo sapiens 18-22 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 0-16 32059452-1 2020 Heme oxygenase-1 (HO-1), an intracellular enzyme that catalyzes the degradation of heme into biliverdin, free iron, and carbon monoxide, exerts anti-inflammatory and cytoprotective effects against endothelial cell injury. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 18-22 32050604-4 2020 However, the expression of hemoxygenase-1 (HO-1) protein was up-regulated upon exposure to floridoside, and two antioxidant enzymes, superoxide dismutase (SOD) and GSH-Px, were activated by floridoside. 2-galactopyranosylglycerol 91-102 heme oxygenase 1 Homo sapiens 27-47 32050604-4 2020 However, the expression of hemoxygenase-1 (HO-1) protein was up-regulated upon exposure to floridoside, and two antioxidant enzymes, superoxide dismutase (SOD) and GSH-Px, were activated by floridoside. Glutathione 164-167 heme oxygenase 1 Homo sapiens 27-47 32050604-4 2020 However, the expression of hemoxygenase-1 (HO-1) protein was up-regulated upon exposure to floridoside, and two antioxidant enzymes, superoxide dismutase (SOD) and GSH-Px, were activated by floridoside. 2-galactopyranosylglycerol 190-201 heme oxygenase 1 Homo sapiens 27-47 31902541-0 2020 Corrigendum to "Cytoprotective effect of Fufang Lurong Jiangu capsule against hydrogen peroxide-induced oxidative stress in bone marrow stromal cell-derived osteoblasts through the Nrf2/HO-1 signaling pathway" [Biomed. Hydrogen Peroxide 78-95 heme oxygenase 1 Homo sapiens 186-190 31848666-0 2020 Heme oxygenase-1 regulates autophagy through carbon-oxygen to alleviate deoxynivalenol-induced hepatic damage. Carbon 45-51 heme oxygenase 1 Homo sapiens 0-16 31848666-0 2020 Heme oxygenase-1 regulates autophagy through carbon-oxygen to alleviate deoxynivalenol-induced hepatic damage. deoxynivalenol 72-86 heme oxygenase 1 Homo sapiens 0-16 31848666-5 2020 Importantly, AAV-mediated liver-specific overexpression of HO-1 reversed DON-induced liver damages, upregulated autophagy and attenuated apoptosis in liver, while AAV-mediated HO-1 silence aggravated DON-induced liver damages, inhibited autophagy and increased apoptosis. deoxynivalenol 73-76 heme oxygenase 1 Homo sapiens 59-63 31848666-5 2020 Importantly, AAV-mediated liver-specific overexpression of HO-1 reversed DON-induced liver damages, upregulated autophagy and attenuated apoptosis in liver, while AAV-mediated HO-1 silence aggravated DON-induced liver damages, inhibited autophagy and increased apoptosis. deoxynivalenol 200-203 heme oxygenase 1 Homo sapiens 59-63 31848666-5 2020 Importantly, AAV-mediated liver-specific overexpression of HO-1 reversed DON-induced liver damages, upregulated autophagy and attenuated apoptosis in liver, while AAV-mediated HO-1 silence aggravated DON-induced liver damages, inhibited autophagy and increased apoptosis. deoxynivalenol 200-203 heme oxygenase 1 Homo sapiens 176-180 31848666-8 2020 Therefore, we suppose that HO-1 might be a potential research target to protect human and animal from liver injuries induced by low dose of DON exposure. deoxynivalenol 140-143 heme oxygenase 1 Homo sapiens 27-31 31787334-5 2020 The sR9-mediated heme oxygenase-1 siRNA (siHO-1) delivery efficiently enhanced apoptotic response to daunorubicin of AML cells and AML-targeted HO-1 silencing improved chemotherapy and prolonged survival in orthotopic myeloid leukemia model. Daunorubicin 101-113 heme oxygenase 1 Homo sapiens 43-47 31705351-10 2020 Overexpression of HO-1 in HER2-expressing SKBR3 breast cancer cells resulted in reduced sensitivity to both pan-HER family kinase inhibitors sapatinib and lapatinib. sapatinib 141-150 heme oxygenase 1 Homo sapiens 18-22 31705351-10 2020 Overexpression of HO-1 in HER2-expressing SKBR3 breast cancer cells resulted in reduced sensitivity to both pan-HER family kinase inhibitors sapatinib and lapatinib. lapatinib 155-164 heme oxygenase 1 Homo sapiens 18-22 31705351-13 2020 Treatment with autophagy inhibitors was able to increase the sensitivity of the HO-1 over-expressing cells to both lapatinib and sapatinib. lapatinib 115-124 heme oxygenase 1 Homo sapiens 80-84 31705351-13 2020 Treatment with autophagy inhibitors was able to increase the sensitivity of the HO-1 over-expressing cells to both lapatinib and sapatinib. sapatinib 129-138 heme oxygenase 1 Homo sapiens 80-84 32141582-5 2020 Furthermore, the expression of antioxidant factor Nrf2, as well as downstream antioxidant genes, including NQO1, SOD1, SOD2, and HO1 was assessed in hydroquinone stimulated ARPE19 cells, in the presence or absence of SAC pretreatment. hydroquinone 149-161 heme oxygenase 1 Homo sapiens 129-132 32010329-8 2020 However, S1PC was found to augment HMOX1 expression, induced by NO donors, such as NOR3. 1-propenylcysteine sulfoxide 9-13 heme oxygenase 1 Homo sapiens 35-40 32096202-12 2020 Melatonin reduced the ROS, MDA, 4-HNE, and 8-OHdG contents but further enhanced the levels of the nuclear factor E2-related factor-2 (Nrf2) and heme oxygenase (HO-1). Melatonin 0-9 heme oxygenase 1 Homo sapiens 160-164 32096202-13 2020 Melatonin-increased viability and melatonin-decreased LDH release were also mediated by the blockage of NF-kappaB or reversed by Nrf2 or HO-1 knockdown. Melatonin 0-9 heme oxygenase 1 Homo sapiens 137-141 32096202-13 2020 Melatonin-increased viability and melatonin-decreased LDH release were also mediated by the blockage of NF-kappaB or reversed by Nrf2 or HO-1 knockdown. Melatonin 34-43 heme oxygenase 1 Homo sapiens 137-141 32096202-16 2020 CONCLUSIONS: Melatonin protected SH SY5Y cells from OGD/R induced oxidative stress, inflammation, apoptosis, and autophagy by blocking NF-kappaB signaling and activating Nrf2/HO-1, Akt, and mTOR/p70S6K/4E-BP-1 pathways, thereby indicating that melatonin is a potential and novel therapeutic drug for ischemic stroke. Melatonin 13-22 heme oxygenase 1 Homo sapiens 175-179 32411446-6 2020 Nrf2/HO-1 also has the ability to modulate calcium levels to prevent ferroptosis, pyroptosis, autophagy, programmed cell necrosis, alkaliptosis, and clockophagy. Calcium 43-50 heme oxygenase 1 Homo sapiens 5-9 32082323-6 2020 Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. Heme 57-61 heme oxygenase 1 Homo sapiens 0-22 32082323-6 2020 Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. Heme 57-61 heme oxygenase 1 Homo sapiens 35-40 32082323-6 2020 Heme oxygenase-1 (HO-1, encoded by HMOX1) produces BV by heme degradation, while biliverdin reductase-A (BLVR-A) generates BR by the subsequent conversion of BV. Bilirubin 123-125 heme oxygenase 1 Homo sapiens 0-22 31347162-0 2020 CD73-dependent adenosine dampens interleukin 1beta-induced CXCL8 production in gingival fibroblasts: Association with heme oxygenase-1 and adenosine monophosphate-activated protein kinase. Adenosine 15-24 heme oxygenase 1 Homo sapiens 118-134 31347162-9 2020 ATP pretreatment impaired IL-1beta-induced CXCL8 secretion and required activation of heme oxygenase-1 (HO-1) and phosphorylated adenosine monophosphate-activated protein kinase (pAMPK). Adenosine Triphosphate 0-3 heme oxygenase 1 Homo sapiens 86-102 31347162-9 2020 ATP pretreatment impaired IL-1beta-induced CXCL8 secretion and required activation of heme oxygenase-1 (HO-1) and phosphorylated adenosine monophosphate-activated protein kinase (pAMPK). Adenosine Triphosphate 0-3 heme oxygenase 1 Homo sapiens 104-108 31347162-11 2020 CONCLUSIONS: CD73-generated adenosine dampens IL-1beta-induced CXCL8 in HGFs and involves HO-1 and pAMPK signaling. Adenosine 28-37 heme oxygenase 1 Homo sapiens 90-94 31974615-5 2020 It was demonstrated that crocetin significantly attenuated the activities of pro-inflammatory cytokines and nuclear factor erythroid-2 related factor 2 (Nrf2)/heme oxygenase-1 signaling. crocetin 25-33 heme oxygenase 1 Homo sapiens 159-175 31884405-0 2020 Protective effect of Hedansanqi Tiaozhi Tang against non-alcoholic fatty liver disease in vitro and in vivo through activating Nrf2/HO-1 antioxidant signaling pathway. hedansanqi 21-31 heme oxygenase 1 Homo sapiens 132-136 31884405-0 2020 Protective effect of Hedansanqi Tiaozhi Tang against non-alcoholic fatty liver disease in vitro and in vivo through activating Nrf2/HO-1 antioxidant signaling pathway. tiaozhi 32-39 heme oxygenase 1 Homo sapiens 132-136 31669540-9 2020 15-Keto PGE2 activated AKT signaling, and the pharmacological AKT inhibitor, LY294002 suppressed the 15-keto PGE2-induced HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 77-85 heme oxygenase 1 Homo sapiens 122-126 32082480-6 2020 Mechanistic studies proved that farrerol could induce an inhibitory phosphorylation of GSK-3beta at Ser9 without affecting the expression level of total GSK-3beta protein and promote the nuclear translocation of Nrf2 as well as the mRNA and protein expression of its downstream target genes heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in EA.hy926 cells. farrerol 32-40 heme oxygenase 1 Homo sapiens 291-307 32064026-4 2020 In piglets fed a DON-contaminated diet, dietary supplementation with BCU significantly decreased the mRNA levels of P70S6K, 4E-BP1, and HSP70 in the liver, the protein expression of HO-1 in the jejunum, and the expression of p-Nrf2 and p-NF-kappaB in the ileum but increased Mn-SOD activity in serum. (3r)-3,4-Dihydro-2h-Chromen-3-Ylmethanol 69-72 heme oxygenase 1 Homo sapiens 182-186 31964423-12 2020 HMOX1 inhibition by tin protoporphyrin (SnPP) in normal DBMSCs resulted in a reduction in proliferation, migration, adhesion, and clone formation processes as compared to the untreated controls. tin protoporphyrin IX 20-38 heme oxygenase 1 Homo sapiens 0-5 31964423-12 2020 HMOX1 inhibition by tin protoporphyrin (SnPP) in normal DBMSCs resulted in a reduction in proliferation, migration, adhesion, and clone formation processes as compared to the untreated controls. tin protoporphyrin IX 40-44 heme oxygenase 1 Homo sapiens 0-5 31964423-13 2020 mRNA and protein analyses of PE-DBMSCs preconditioned with H2O2 at lower doses showed upregulation of HMOX1 expression. Water 59-63 heme oxygenase 1 Homo sapiens 102-107 32065759-7 2020 Hyperoxia was also found to diminish DNA damage and generation of free radicals initiated in B[a]P-treated cells which was attributed to an significant rise of Nrf2, leading to elevated antioxidant activities or detoxification proteins including heme oxygenase 1 (HO-1), superoxide dismutase (SOD), glutathione peroxidase-1/2 (GPX-1/2), CAT, GST and glutathione (GSH). Phosphorus 97-98 heme oxygenase 1 Homo sapiens 246-262 32065759-7 2020 Hyperoxia was also found to diminish DNA damage and generation of free radicals initiated in B[a]P-treated cells which was attributed to an significant rise of Nrf2, leading to elevated antioxidant activities or detoxification proteins including heme oxygenase 1 (HO-1), superoxide dismutase (SOD), glutathione peroxidase-1/2 (GPX-1/2), CAT, GST and glutathione (GSH). Phosphorus 97-98 heme oxygenase 1 Homo sapiens 264-268 32082188-3 2020 This review aims to provide an up-to-date and critical overview of the molecular mechanisms by which bilirubin derived from plasma or from HMOX1 activation in vascular cells affects endothelial function. Bilirubin 101-110 heme oxygenase 1 Homo sapiens 139-144 32082188-5 2020 In this context, therapeutic interventions aimed at mildly increasing serum bilirubin as well as bilirubin generated endogenously by endothelial HMOX1 should be considered. Bilirubin 97-106 heme oxygenase 1 Homo sapiens 145-150 31991717-4 2020 In our experimental model, main carnosine beneficial effects were: (1) the modulation of nitric oxide production and metabolism; (2) the amelioration of the macrophage energy state; (3) the decrease of the expressions of pro-oxidant enzymes (Nox-2, Cox-2) and of the lipid peroxidation product malondialdehyde; (4) the restoration and/or increase of the expressions of antioxidant enzymes (Gpx1, SOD-2 and Cat); (5) the increase of the transforming growth factor-beta1 (TGF-beta1) and the down-regulation of the expressions of interleukins 1beta and 6 (IL-1beta and IL-6) and 6) the increase of the expressions of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase-1 (HO-1). Carnosine 32-41 heme oxygenase 1 Homo sapiens 669-685 32020857-12 2020 CONCLUSIONS: The neuroprotective effects of Danshensu on rotenone-induced PD are attributed to the anti-oxidative properties by activating PI3K/AKT/Nrf2 pathway and increasing Nrf2-induced expression of HO-1, GCLC, and GCLM, at least in part. 3,4-dihydroxyphenyllactic acid 44-53 heme oxygenase 1 Homo sapiens 203-207 32020857-12 2020 CONCLUSIONS: The neuroprotective effects of Danshensu on rotenone-induced PD are attributed to the anti-oxidative properties by activating PI3K/AKT/Nrf2 pathway and increasing Nrf2-induced expression of HO-1, GCLC, and GCLM, at least in part. Rotenone 57-65 heme oxygenase 1 Homo sapiens 203-207 31974389-3 2020 We reported that both polyphenols show antioxidant and oto-protective activity in the cochlea by up-regulating Nrf-2/HO-1 pathway and downregulating p53 phosphorylation. Polyphenols 22-33 heme oxygenase 1 Homo sapiens 117-121 31669540-0 2020 15-Keto prostaglandin E2 induces heme oxygenase-1 expression through activation of Nrf2 in human colon epithelial CCD 841 CoN cells. 15-ketoprostaglandin E2 0-24 heme oxygenase 1 Homo sapiens 33-49 31669540-5 2020 In this study, we found that 15-keto PGE2 upregulated the expression of heme oxygenase-1 (HO-1), a representative antioxidative and anti-inflammatory enzyme, at both transcriptional and translational levels, in human colon epithelial CCD 841 CoN cells. Dinoprostone 37-41 heme oxygenase 1 Homo sapiens 72-88 31669540-5 2020 In this study, we found that 15-keto PGE2 upregulated the expression of heme oxygenase-1 (HO-1), a representative antioxidative and anti-inflammatory enzyme, at both transcriptional and translational levels, in human colon epithelial CCD 841 CoN cells. Dinoprostone 37-41 heme oxygenase 1 Homo sapiens 90-94 31669540-8 2020 Furthermore, the silencing of the Nrf2 gene abolished 15-keto PGE2-induced HO-1 expression in CCD 841 CoN cells. 15-ketoprostaglandin E2 54-66 heme oxygenase 1 Homo sapiens 75-79 31669540-9 2020 15-Keto PGE2 activated AKT signaling, and the pharmacological AKT inhibitor, LY294002 suppressed the 15-keto PGE2-induced HO-1 expression. 15-ketoprostaglandin E2 0-12 heme oxygenase 1 Homo sapiens 122-126 31669540-9 2020 15-Keto PGE2 activated AKT signaling, and the pharmacological AKT inhibitor, LY294002 suppressed the 15-keto PGE2-induced HO-1 expression. 15-ketoprostaglandin E2 101-113 heme oxygenase 1 Homo sapiens 122-126 31669540-11 2020 N-acetyl-l-cysteine treatment attenuated the 15-keto PGE2-induced phosphorylation of GSK3beta, transcriptional activity of Nrf2, and subsequently HO-1 expression. Acetylcysteine 0-19 heme oxygenase 1 Homo sapiens 146-150 31669540-11 2020 N-acetyl-l-cysteine treatment attenuated the 15-keto PGE2-induced phosphorylation of GSK3beta, transcriptional activity of Nrf2, and subsequently HO-1 expression. Dinoprostone 53-57 heme oxygenase 1 Homo sapiens 146-150 31669540-13 2020 In conclusion, 15-keto PGE2 induces HO-1 expression through Nrf2 activation in human colon epithelial cells. 15-ketoprostaglandin E2 15-27 heme oxygenase 1 Homo sapiens 36-40 31952230-10 2020 CONCLUSIONS: MVS-induced HO-1 is, at least in part, mediated through a p47phox/Nox2/ROS-dependent activation of c-Src/PDGFRalpha/PI3K/Akt-regulated Nrf2/ARE axis and suppresses the TNF-alpha-mediated inflammatory responses in HPAEpiCs. mevastatin 13-16 heme oxygenase 1 Homo sapiens 25-29 31952230-0 2020 Induction of HO-1 by Mevastatin Mediated via a Nox/ROS-Dependent c-Src/PDGFRalpha/PI3K/Akt/Nrf2/ARE Cascade Suppresses TNF-alpha-Induced Lung Inflammation. mevastatin 21-31 heme oxygenase 1 Homo sapiens 13-17 31677563-8 2020 In the duodenum, glyphosate remarkably increased the relative mRNA expression levels of Nrf2 (linear and quadratic, P < 0.05) and NQO1 (linear and quadratic, P < 0.05) and reduced the relative mRNA expression levels of GPx1, HO-1 and GCLM (linear and quadratic, P < 0.05). glyphosate 17-27 heme oxygenase 1 Homo sapiens 231-235 32668967-2 2020 The aim of the this study was to examine the effects of [Formula: see text] (a rare protopanaxatriol-type ginsenoside fraction; Rg2, Rg4, Rg6, Rh1, and Rh4) on heme oxygenase-1 (HO-1) induction and on the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-)2 in lipopolysaccharide (LPS)-activated human pulmonary artery endothelial cells (HPAECs). protopanaxatriol 84-100 heme oxygenase 1 Homo sapiens 160-176 31952230-1 2020 BACKGROUND: Mevastatin (MVS), a 3-hydroxy-3-methylglutaryl coenzyme, a reductase (HMG-CoA) inhibitor, has anti-inflammatory effects potentially via up-regulation of heme oxygenase-1 (HO-1). mevastatin 12-22 heme oxygenase 1 Homo sapiens 165-181 31952230-1 2020 BACKGROUND: Mevastatin (MVS), a 3-hydroxy-3-methylglutaryl coenzyme, a reductase (HMG-CoA) inhibitor, has anti-inflammatory effects potentially via up-regulation of heme oxygenase-1 (HO-1). mevastatin 12-22 heme oxygenase 1 Homo sapiens 183-187 31952230-1 2020 BACKGROUND: Mevastatin (MVS), a 3-hydroxy-3-methylglutaryl coenzyme, a reductase (HMG-CoA) inhibitor, has anti-inflammatory effects potentially via up-regulation of heme oxygenase-1 (HO-1). mevastatin 24-27 heme oxygenase 1 Homo sapiens 165-181 31952230-1 2020 BACKGROUND: Mevastatin (MVS), a 3-hydroxy-3-methylglutaryl coenzyme, a reductase (HMG-CoA) inhibitor, has anti-inflammatory effects potentially via up-regulation of heme oxygenase-1 (HO-1). mevastatin 24-27 heme oxygenase 1 Homo sapiens 183-187 31952230-2 2020 However, the mechanisms underlying MVS-induced HO-1 expression remain largely unknown in human pulmonary alveolar epithelial cells (HPAEpiCs). mevastatin 35-38 heme oxygenase 1 Homo sapiens 47-51 31952230-5 2020 RESULTS: Upregulation of HO-1 by MVS attenuated the tumor necrosis factor (TNF)-alpha-stimulated ICAM-1 expression associated with THP-1 adhesion to HPAEpiCs. mevastatin 33-36 heme oxygenase 1 Homo sapiens 25-29 31952230-6 2020 These inhibitory effects of HO-1 were reversed by tin protoporphyrin (SnPP)IX or by transfection with HO-1 siRNA. tin protoporphyrin IX 50-68 heme oxygenase 1 Homo sapiens 28-32 31952230-6 2020 These inhibitory effects of HO-1 were reversed by tin protoporphyrin (SnPP)IX or by transfection with HO-1 siRNA. tin protoporphyrin IX 70-74 heme oxygenase 1 Homo sapiens 28-32 31952230-7 2020 MVS-induced HO-1 expression was mediated via NADPH oxidase (Nox)-derived reactive oxygen species (ROS) generation. mevastatin 0-3 heme oxygenase 1 Homo sapiens 12-16 31952230-7 2020 MVS-induced HO-1 expression was mediated via NADPH oxidase (Nox)-derived reactive oxygen species (ROS) generation. NADP 45-50 heme oxygenase 1 Homo sapiens 12-16 31952230-7 2020 MVS-induced HO-1 expression was mediated via NADPH oxidase (Nox)-derived reactive oxygen species (ROS) generation. Oxygen 82-88 heme oxygenase 1 Homo sapiens 12-16 31678598-8 2020 Activated AMPKalpha induced activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), which are proteins functioning in redox balance. ampkalpha 10-19 heme oxygenase 1 Homo sapiens 97-113 31678598-8 2020 Activated AMPKalpha induced activation of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), which are proteins functioning in redox balance. ampkalpha 10-19 heme oxygenase 1 Homo sapiens 115-119 32668967-2 2020 The aim of the this study was to examine the effects of [Formula: see text] (a rare protopanaxatriol-type ginsenoside fraction; Rg2, Rg4, Rg6, Rh1, and Rh4) on heme oxygenase-1 (HO-1) induction and on the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase (COX-)2 in lipopolysaccharide (LPS)-activated human pulmonary artery endothelial cells (HPAECs). Ginsenosides 106-117 heme oxygenase 1 Homo sapiens 160-176 31764154-10 2020 Blockade of lipoxin A4 production in mesenchymal stromal cells, and of prostaglandin E2 synthesis in mesenchymal stromal cell/macrophage cocultures, prevented upregulation of heme oxygenase-1 in macrophages (from 9.6 +- 5.5-fold to 2.3 +- 1.3 and 2.4 +- 2.3 respectively, P = 0.004). lipoxin A4 12-22 heme oxygenase 1 Homo sapiens 175-191 31796664-6 2020 The following clusters were found in the dendrogram: Nrf2 and p21 with ATP5B and GADPH in all the tissues and with NRF1 in all except the tumor tissues with metastasis; Bach1 with ATP5B and GAPDH in the tumor tissues; Keap1 with p62 in all the tissues, with LC3 in the tumor tissues and with NRF1 and HO1 in the tumor tissues with metastasis. gadph 81-86 heme oxygenase 1 Homo sapiens 301-304 33268671-0 2020 (-)-Epigallocatechin-3-gallate inhibits RANKL-induced osteoclastogenesis via downregulation of NFATc1 and suppression of HO-1-HMGB1-RAGE pathway. epigallocatechin gallate 0-30 heme oxygenase 1 Homo sapiens 121-125 33268671-8 2020 We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). epigallocatechin gallate 19-23 heme oxygenase 1 Homo sapiens 36-52 33268671-8 2020 We also found that EGCG upregulated heme oxygenase-1 (HO-1) and suppressed the extracellular release of high-mobility group box 1 (HMGB1). epigallocatechin gallate 19-23 heme oxygenase 1 Homo sapiens 54-58 33268671-10 2020 In summary, our study showed that EGCG could inhibit osteoclast differentiation through the downregulation of NFATc1 and the suppression of the HO-1-HMGB1-RAGE pathway. epigallocatechin gallate 34-38 heme oxygenase 1 Homo sapiens 144-148 31646664-4 2020 The dose-dependent protective effects against H2 O2 -induced oxidative stress of the rutin-Lys MRPs may involve the inhibition of reactive oxygen species generation, enhancement of the superoxide dismutase and catalase activities, along with the activation of the Nrf2-dependent pathway and upregulation of phase II antioxidant genes (including NQO1, HO-1, GCLG, and GCLM). Water 46-51 heme oxygenase 1 Homo sapiens 351-355 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Heme 88-92 heme oxygenase 1 Homo sapiens 0-16 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Heme 88-92 heme oxygenase 1 Homo sapiens 18-22 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. ferrous iron 101-113 heme oxygenase 1 Homo sapiens 0-16 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. ferrous iron 101-113 heme oxygenase 1 Homo sapiens 18-22 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 0-16 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 18-22 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 0-16 33016915-4 2020 Heme oxygenase 1 (HO-1) is a 32 kDa enzyme, which catalyzes the degradation of cellular heme to free ferrous iron, biliverdin, and carbon monoxide under stressful conditions. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 18-22 33016915-6 2020 The beneficial roles of HO-1 overexpression in AD brains are widely accepted due to its ability to convert pro-oxidant heme to biliverdin and bilirubin (antioxidants), which promote restoration of a suitable tissue redox microenvironment. Heme 119-123 heme oxygenase 1 Homo sapiens 24-28 33016915-6 2020 The beneficial roles of HO-1 overexpression in AD brains are widely accepted due to its ability to convert pro-oxidant heme to biliverdin and bilirubin (antioxidants), which promote restoration of a suitable tissue redox microenvironment. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 24-28 33016915-6 2020 The beneficial roles of HO-1 overexpression in AD brains are widely accepted due to its ability to convert pro-oxidant heme to biliverdin and bilirubin (antioxidants), which promote restoration of a suitable tissue redox microenvironment. Bilirubin 142-151 heme oxygenase 1 Homo sapiens 24-28 32047536-4 2020 Furthermore, Nrf2 downstream antioxidant genes such as HO-1(heme oxygenase 1) and NQO1 (NAD (P) H: quinone oxidoreductase 1) were upregulated in SKOV3/DDP cells with VK3 treatment, which indicated VK3 activated Nrf2 signaling in SKOV3/DDP cells. Vitamin K 166-169 heme oxygenase 1 Homo sapiens 55-59 32047536-4 2020 Furthermore, Nrf2 downstream antioxidant genes such as HO-1(heme oxygenase 1) and NQO1 (NAD (P) H: quinone oxidoreductase 1) were upregulated in SKOV3/DDP cells with VK3 treatment, which indicated VK3 activated Nrf2 signaling in SKOV3/DDP cells. Vitamin K 166-169 heme oxygenase 1 Homo sapiens 60-76 32047536-4 2020 Furthermore, Nrf2 downstream antioxidant genes such as HO-1(heme oxygenase 1) and NQO1 (NAD (P) H: quinone oxidoreductase 1) were upregulated in SKOV3/DDP cells with VK3 treatment, which indicated VK3 activated Nrf2 signaling in SKOV3/DDP cells. Vitamin K 197-200 heme oxygenase 1 Homo sapiens 55-59 32047536-4 2020 Furthermore, Nrf2 downstream antioxidant genes such as HO-1(heme oxygenase 1) and NQO1 (NAD (P) H: quinone oxidoreductase 1) were upregulated in SKOV3/DDP cells with VK3 treatment, which indicated VK3 activated Nrf2 signaling in SKOV3/DDP cells. Vitamin K 197-200 heme oxygenase 1 Homo sapiens 60-76 31646664-4 2020 The dose-dependent protective effects against H2 O2 -induced oxidative stress of the rutin-Lys MRPs may involve the inhibition of reactive oxygen species generation, enhancement of the superoxide dismutase and catalase activities, along with the activation of the Nrf2-dependent pathway and upregulation of phase II antioxidant genes (including NQO1, HO-1, GCLG, and GCLM). Rutin 85-90 heme oxygenase 1 Homo sapiens 351-355 31646664-4 2020 The dose-dependent protective effects against H2 O2 -induced oxidative stress of the rutin-Lys MRPs may involve the inhibition of reactive oxygen species generation, enhancement of the superoxide dismutase and catalase activities, along with the activation of the Nrf2-dependent pathway and upregulation of phase II antioxidant genes (including NQO1, HO-1, GCLG, and GCLM). pentalysine 91-94 heme oxygenase 1 Homo sapiens 351-355 31882813-5 2019 Besides, Mn-TAT PTD-Ngb activated the phosphoinositide-3 kinase (PI3K)/Akt signaling pathway, which up-regulated the expression of nuclear factor E2-related factor 2 (Nrf2), Heme oxygenase-1 (HO-1), superoxide dismutase (SOD), catalase (CAT). Superoxides 199-209 heme oxygenase 1 Homo sapiens 174-190 31411335-1 2020 Objectives The number of dinucleotide repeats (GT) nmodulate expression of heme oxygenase 1 (HMOX1), a stress response gene. Dinucleoside Phosphates 25-37 heme oxygenase 1 Homo sapiens 75-91 31411335-1 2020 Objectives The number of dinucleotide repeats (GT) nmodulate expression of heme oxygenase 1 (HMOX1), a stress response gene. Dinucleoside Phosphates 25-37 heme oxygenase 1 Homo sapiens 93-98 31895879-0 2020 Berberine Attenuates Cholesterol Accumulation in Macrophage Foam Cells by Suppressing AP-1 Activity and Activation of the Nrf2/HO-1 Pathway. Berberine 0-9 heme oxygenase 1 Homo sapiens 127-131 31895879-8 2020 Mechanisms study showed that berberine can suppress scavenger receptor expression via inhibiting the activity of AP-1 and upregulate ATP-binding cassette transporter via activating Nrf2/HO-1 signaling in human macrophage. Berberine 29-38 heme oxygenase 1 Homo sapiens 186-190 31865348-11 2020 In addition, EX4 markedly increased expression of NRF2, HO-1, and NQO-1 and significantly improved protein expression of NRF2 and HO-1 in H2O2-treated ARPE-19 cells, caused by increased nuclear NRF2 protein expression. Water 138-142 heme oxygenase 1 Homo sapiens 130-134 31518892-5 2020 In addition, CBD induced the expression of several NRF2 target genes, with heme oxygenase 1 (HMOX1) being the gene and the protein most upregulated by CBD. Cannabidiol 13-16 heme oxygenase 1 Homo sapiens 75-91 31518892-5 2020 In addition, CBD induced the expression of several NRF2 target genes, with heme oxygenase 1 (HMOX1) being the gene and the protein most upregulated by CBD. Cannabidiol 13-16 heme oxygenase 1 Homo sapiens 93-98 31518892-5 2020 In addition, CBD induced the expression of several NRF2 target genes, with heme oxygenase 1 (HMOX1) being the gene and the protein most upregulated by CBD. Cannabidiol 151-154 heme oxygenase 1 Homo sapiens 75-91 31518892-5 2020 In addition, CBD induced the expression of several NRF2 target genes, with heme oxygenase 1 (HMOX1) being the gene and the protein most upregulated by CBD. Cannabidiol 151-154 heme oxygenase 1 Homo sapiens 93-98 31810001-7 2020 HO-1 attenuated reactive oxygen species induced by H2O2 or pyocyanin treatment in PC-3 and DU145 cells. Oxygen 25-31 heme oxygenase 1 Homo sapiens 0-4 31810001-7 2020 HO-1 attenuated reactive oxygen species induced by H2O2 or pyocyanin treatment in PC-3 and DU145 cells. Water 51-55 heme oxygenase 1 Homo sapiens 0-4 31810001-7 2020 HO-1 attenuated reactive oxygen species induced by H2O2 or pyocyanin treatment in PC-3 and DU145 cells. Pyocyanine 59-68 heme oxygenase 1 Homo sapiens 0-4 31810001-8 2020 HO-1 further reduced PC-3 and DU145 cell apoptosis induced by H2O2 or serum starvation. Water 62-66 heme oxygenase 1 Homo sapiens 0-4 31877176-0 2019 alpha-Lipoic acid prevents against cisplatin cytotoxicity via activation of the NRF2/HO-1 antioxidant pathway. Thioctic Acid 0-17 heme oxygenase 1 Homo sapiens 85-89 31877176-0 2019 alpha-Lipoic acid prevents against cisplatin cytotoxicity via activation of the NRF2/HO-1 antioxidant pathway. Cisplatin 35-44 heme oxygenase 1 Homo sapiens 85-89 31882813-5 2019 Besides, Mn-TAT PTD-Ngb activated the phosphoinositide-3 kinase (PI3K)/Akt signaling pathway, which up-regulated the expression of nuclear factor E2-related factor 2 (Nrf2), Heme oxygenase-1 (HO-1), superoxide dismutase (SOD), catalase (CAT). Superoxides 199-209 heme oxygenase 1 Homo sapiens 192-196 31614142-6 2019 Sofalcone induced HO-1 protein expression, which was dependent on increased nuclear accumulation of Nrf2 in human colon carcinoma cells. sofalcone 0-9 heme oxygenase 1 Homo sapiens 18-22 31878134-6 2019 Moreover, Nrf2 and its downstream heme oxygenase 1 (HO-1) were inhibited by Cd exposure, and Kelch-like ECH-associated protein 1 (Keap1), the inhibitory protein of Nrf2, was increased. Cadmium 76-78 heme oxygenase 1 Homo sapiens 34-50 31878134-6 2019 Moreover, Nrf2 and its downstream heme oxygenase 1 (HO-1) were inhibited by Cd exposure, and Kelch-like ECH-associated protein 1 (Keap1), the inhibitory protein of Nrf2, was increased. Cadmium 76-78 heme oxygenase 1 Homo sapiens 52-56 31878134-8 2019 In conclusion, the inhibition of Nrf2, HO-1, and the activation of NF-kappaB, NLRP3, and MAPKs all contribute to Cd-induced liver injury. Cadmium 113-115 heme oxygenase 1 Homo sapiens 39-43 31729530-0 2019 Moracin attenuates LPS-induced inflammation in nucleus pulposus cells via Nrf2/HO-1 and NF-kappaB/TGF-beta pathway. moracin C 0-7 heme oxygenase 1 Homo sapiens 79-83 31729530-7 2019 Together, our results demonstrated the ability of moracin to antagonize LPS-mediated inflammation in primary culture of nucleus pulposus in intervertebral disc by partly regulating the Nrf2/HO-1 and NF-kappaB/TGF-beta pathway in nucleus pulposus cells. moracin C 50-57 heme oxygenase 1 Homo sapiens 190-194 31861550-4 2019 The TU-inducible transcription of Nrf1 and Nrf2, as well as GCLM (glutamate cysteine ligase modifier subunit) and HO-1 (heme oxygenase 1), was accompanied by activation of ER stress signaling networks. Tunicamycin 4-6 heme oxygenase 1 Homo sapiens 66-136 31861550-9 2019 Furthermore,&nbsp;caNrf2DeltaN also enhanced induction of PERK and IRE1 by TU, but reduced expression of ATF4 and HO-1. Tunicamycin 79-81 heme oxygenase 1 Homo sapiens 118-122 31614142-13 2019 The protective effects of sofalcone against colon damage and inflammation were significantly inhibited by co-administration of an HO-1 inhibitor. sofalcone 26-35 heme oxygenase 1 Homo sapiens 130-134 31614142-14 2019 In conclusion, sofalcone activated the Nrf2-HO-1 pathway by covalently binding to KEAP1 via Michael addition, and may confer anti-colitic effects by inducing Nrf2 activation. sofalcone 15-24 heme oxygenase 1 Homo sapiens 44-48 31659617-0 2019 Grape Seed Procyanidins Attenuates Cisplatin-induced Human Embryonic Renal Cell Cytotoxicity by Modulating Heme Oxygenase-1 in Vitro. procyanidin 11-23 heme oxygenase 1 Homo sapiens 107-123 31704097-4 2019 BVR-deficiency induces the activity of Nrf2 transcription factor and increases heme oxygenase-1 (HO-1) level, which is accompanied by the reduction of cellular heme content, increase in a free iron fraction and oxidative stress. Heme 79-83 heme oxygenase 1 Homo sapiens 97-101 31704097-5 2019 Accordingly, the phenotype of BVR-deficient cells can be mimicked by hemin or iron overload, whereas depletion of HO-1 in BVR-deficient ECs abrogates the increase in intracellular free iron and oxidative stress, preventing the loss of endothelial markers. Iron 185-189 heme oxygenase 1 Homo sapiens 114-118 31704097-8 2019 Collectively, the non-enzymatic activity of BVR contributes to the maintenance of healthy endothelial phenotype through the prevention of HO-1-dependent iron-overload, oxidative stress and subsequent endothelial-to-mesenchymal transition (EndMT). Iron 153-157 heme oxygenase 1 Homo sapiens 138-142 31847280-12 2019 DHA supplementation increased (p < 0.05) mRNA expression of Nrf2, HO-1, GPx1, and CAT in the ID group. artenimol 0-3 heme oxygenase 1 Homo sapiens 69-73 31847280-13 2019 In addition, the protein expression of Nrf2 was downregulated (p < 0.05) in the liver of IUGR-affected piglets and DHA supplementation increased (p < 0.05) the protein content of Nrf2 and HO-1. artenimol 118-121 heme oxygenase 1 Homo sapiens 194-198 31817533-0 2019 Dietary Curcumin Supplementation Increases Antioxidant Capacity, Upregulates Nrf2 and Hmox1 Levels in the Liver of Piglet Model with Intrauterine Growth Retardation. Curcumin 8-16 heme oxygenase 1 Homo sapiens 86-91 31817533-8 2019 Dietary curcumin supplementation increased body-weight gain, feed intake, activities of antioxidant enzymes, and the expressions of nuclear factor, erythroid 2-like 2 (Nrf2) and heme oxygenase-1 (Hmox1) proteins in the liver of weaned piglets with IUGR. Curcumin 8-16 heme oxygenase 1 Homo sapiens 178-194 31817533-8 2019 Dietary curcumin supplementation increased body-weight gain, feed intake, activities of antioxidant enzymes, and the expressions of nuclear factor, erythroid 2-like 2 (Nrf2) and heme oxygenase-1 (Hmox1) proteins in the liver of weaned piglets with IUGR. Curcumin 8-16 heme oxygenase 1 Homo sapiens 196-201 31817533-10 2019 Curcumin could efficiently improve the growth, increase hepatic antioxidant capacity, and upregulate Nrf2 and Hmox1 levels in the liver of IUGR weaned piglets. Curcumin 0-8 heme oxygenase 1 Homo sapiens 110-115 31885818-0 2019 FA-97, a New Synthetic Caffeic Acid Phenethyl Ester Derivative, Protects against Oxidative Stress-Mediated Neuronal Cell Apoptosis and Scopolamine-Induced Cognitive Impairment by Activating Nrf2/HO-1 Signaling. fa-97 0-5 heme oxygenase 1 Homo sapiens 195-199 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Heme 34-38 heme oxygenase 1 Homo sapiens 0-16 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Heme 34-38 heme oxygenase 1 Homo sapiens 18-22 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Biliverdine 63-81 heme oxygenase 1 Homo sapiens 0-16 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Biliverdine 63-81 heme oxygenase 1 Homo sapiens 18-22 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 0-16 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 18-22 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 100-102 heme oxygenase 1 Homo sapiens 0-16 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 100-102 heme oxygenase 1 Homo sapiens 18-22 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Iron 109-113 heme oxygenase 1 Homo sapiens 0-16 31704095-1 2019 Heme oxygenase-1 (HO-1) catalyzes heme degradation to generate biliverdin-IXalpha, carbon monoxide (CO), and iron. Iron 109-113 heme oxygenase 1 Homo sapiens 18-22 31704095-2 2019 The HO-1/CO system confers cytoprotection in animal models of organ injury and disease, via modulation of inflammation and apoptosis. Carbon Monoxide 9-11 heme oxygenase 1 Homo sapiens 4-8 31921135-2 2019 Based on studies showing the potential role of heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme and has anti-inflammatory properties in SLE development, we decided to explore HO-1 in LN. Heme 47-51 heme oxygenase 1 Homo sapiens 65-69 31159593-8 2019 We found that H2O2 stimulation-evoked HUVECs oxidative damage meanwhile impeded HO-1 expression. Hydrogen Peroxide 14-18 heme oxygenase 1 Homo sapiens 80-84 30825052-9 2019 Additionally, it was determined that Pyr-1 generates oxidative and proteotoxic stress by provoking the accumulation of ROS, resulting in the overexpression of the stress-related hmox-1 mRNA transcripts and protein and a marked increase in poly-ubiquitinated proteins. Pyr-1 37-42 heme oxygenase 1 Homo sapiens 178-184 30825052-9 2019 Additionally, it was determined that Pyr-1 generates oxidative and proteotoxic stress by provoking the accumulation of ROS, resulting in the overexpression of the stress-related hmox-1 mRNA transcripts and protein and a marked increase in poly-ubiquitinated proteins. Reactive Oxygen Species 119-122 heme oxygenase 1 Homo sapiens 178-184 31659617-3 2019 This study reported whether O-GSP can antagonize the cisplatin-induced cytotoxicity in HEK293 cells through inducing HO-1 protein expression. GSP-6 glycosulfopeptide 28-33 heme oxygenase 1 Homo sapiens 117-121 31659617-3 2019 This study reported whether O-GSP can antagonize the cisplatin-induced cytotoxicity in HEK293 cells through inducing HO-1 protein expression. Cisplatin 53-62 heme oxygenase 1 Homo sapiens 117-121 31659617-5 2019 Herein, We found that O-GSP can antagonize cisplatin nephrotoxicity through regulating the expression of HO-1. GSP-6 glycosulfopeptide 22-27 heme oxygenase 1 Homo sapiens 105-109 31659617-5 2019 Herein, We found that O-GSP can antagonize cisplatin nephrotoxicity through regulating the expression of HO-1. Cisplatin 43-52 heme oxygenase 1 Homo sapiens 105-109 31659617-7 2019 Interestingly, p38 MAPK plays a major role in HO-1 expression induced by O-GSP. GSP-6 glycosulfopeptide 73-78 heme oxygenase 1 Homo sapiens 46-50 31659617-8 2019 And O-GSP can modulate the decrease of Nrf2 and HO-1 expression induced by cisplatin, and improve the cisplatin-induced activity and apoptosis rate of cells by stimulating the expression of HO-1. GSP-6 glycosulfopeptide 4-9 heme oxygenase 1 Homo sapiens 48-52 31659617-8 2019 And O-GSP can modulate the decrease of Nrf2 and HO-1 expression induced by cisplatin, and improve the cisplatin-induced activity and apoptosis rate of cells by stimulating the expression of HO-1. GSP-6 glycosulfopeptide 4-9 heme oxygenase 1 Homo sapiens 190-194 31659617-8 2019 And O-GSP can modulate the decrease of Nrf2 and HO-1 expression induced by cisplatin, and improve the cisplatin-induced activity and apoptosis rate of cells by stimulating the expression of HO-1. Cisplatin 75-84 heme oxygenase 1 Homo sapiens 48-52 31659617-8 2019 And O-GSP can modulate the decrease of Nrf2 and HO-1 expression induced by cisplatin, and improve the cisplatin-induced activity and apoptosis rate of cells by stimulating the expression of HO-1. Cisplatin 102-111 heme oxygenase 1 Homo sapiens 190-194 31659617-10 2019 Collectively, the results indicated that O-GSP induced the expression of HO-1 through p38MAPK and Nrf2 pathway in HEK293 cells. GSP-6 glycosulfopeptide 41-46 heme oxygenase 1 Homo sapiens 73-77 31659617-0 2019 Grape Seed Procyanidins Attenuates Cisplatin-induced Human Embryonic Renal Cell Cytotoxicity by Modulating Heme Oxygenase-1 in Vitro. Cisplatin 35-44 heme oxygenase 1 Homo sapiens 107-123 31556759-11 2019 AX-SLN reduced the p-AKT which is accountable for the reduction in the NF-kB expression and also reduced the expression of Keap1 and NF-kB along with increasing the expression of HO-1 and Nrf-2. Amoxicillin 0-2 heme oxygenase 1 Homo sapiens 179-183 30535793-6 2019 Treatment with NAC significantly increased jejunal superoxide dismutase activity, reduced glutathione: oxidized glutathione ratio, and the mRNA abundance of nuclear respiratory factor 2, heme oxygenase 1, and superoxide dismutase 2 in the IUGR-NAC piglets compared with the IUGR-CON piglets. Acetylcysteine 15-18 heme oxygenase 1 Homo sapiens 187-203 31347718-5 2019 Fortunately, OS preconditioning can increase the expression of superoxide dismutase, catalase, NQO1, and heme oxygenase 1 through the nuclear factor erythroid 2-related factor 2 pathway, thereby decreased the intracellular reactive oxygen species (ROS) levels, relieved the damage of ROS to mitochondria, DNA and cell membrane, enhanced the anti-OS ability of BMSCs, and promoted the survival of BMSCs under OS. Reactive Oxygen Species 223-246 heme oxygenase 1 Homo sapiens 105-121 31950025-0 2019 Erratum: 15-Deoxy-Delta12,14-prostaglandin J2 Upregulates the Expression of 15-Hydroxyprostaglandin Dehydrogenase by Inducing AP-1 Activation and Heme Oxygenase-1 Expression in Human Colon Cancer Cells. 15-deoxy-delta(12,14)-prostaglandin J2 9-45 heme oxygenase 1 Homo sapiens 146-162 31734545-7 2019 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase and heme oxygenase-1 and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 21-25 heme oxygenase 1 Homo sapiens 147-163 31734545-7 2019 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase and heme oxygenase-1 and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 147-163 31734545-7 2019 We hypothesized that PpIX fluorescence intensity results from the interplay between the metabolic clearance of PpIX mediated by ferrochelatase and heme oxygenase-1 and the cellular efflux of PpIX through the ATP-binding cassette subfamily G member 2 (ABCG2). protoporphyrin IX 111-115 heme oxygenase 1 Homo sapiens 147-163 31559456-11 2019 CONCLUSION: The cytotoxic effects of CDDP and THP were both enhanced under HO-1 knockdown conditions as well as under conditions that inhibit the activation pathway of HO-1 by EGFR inhibitors. pirarubicin 46-49 heme oxygenase 1 Homo sapiens 75-79 31351398-9 2019 RESULTS: HaCAT cells treated with 40 muM CoCl2 could not only reduce cell viability, promote cell apoptosis, arrest G1 phase of cell cycle and increase the activity of reactive oxygen species but also downregulate the expressions of c-myc, c-fos, transforming growth factor-alpha, Nrf2, heme oxygenase-1, and gamma-glutamyl cysteine synthetase. cobaltous chloride 41-46 heme oxygenase 1 Homo sapiens 287-303 31404677-0 2019 Photoirradiation after aminolevulinic acid treatment suppresses cancer cell proliferation through the HO-1/p21 pathway. Aminolevulinic Acid 23-42 heme oxygenase 1 Homo sapiens 102-106 31404677-8 2019 RESULTS: mRNAs of PAI-1, HO-1, and p21 were upregulated after photoirradiation of HEK293, which was suppressed by N-acetyl-L-cysteine, a reactive oxygen species (ROS) scavenger. Acetylcysteine 114-133 heme oxygenase 1 Homo sapiens 25-29 31661135-7 2019 In the present analysis, HMOX1 and HSPA1A were identified as the most highly upregulated genes following sulforaphane treatment, suggesting their potential value as biomarkers to guide clinical trials. sulforaphane 105-117 heme oxygenase 1 Homo sapiens 25-30 31559456-0 2019 Epidermal growth factor receptor/heme oxygenase-1 axis is involved in chemoresistance to cisplatin and pirarubicin in HepG2 cell lines and hepatoblastoma specimens. Cisplatin 89-98 heme oxygenase 1 Homo sapiens 33-49 31559456-0 2019 Epidermal growth factor receptor/heme oxygenase-1 axis is involved in chemoresistance to cisplatin and pirarubicin in HepG2 cell lines and hepatoblastoma specimens. pirarubicin 103-114 heme oxygenase 1 Homo sapiens 33-49 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. Cisplatin 153-162 heme oxygenase 1 Homo sapiens 76-92 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. Cisplatin 153-162 heme oxygenase 1 Homo sapiens 94-98 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. pirarubicin 167-178 heme oxygenase 1 Homo sapiens 76-92 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. pirarubicin 167-178 heme oxygenase 1 Homo sapiens 94-98 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. cita 180-184 heme oxygenase 1 Homo sapiens 76-92 31559456-1 2019 PURPOSE: To investigate the possibility that the antioxidant stress protein Heme oxygenase-1 (HO-1) is involved in the acquisition of chemoresistance in cisplatin and pirarubicin (CITA) therapy. cita 180-184 heme oxygenase 1 Homo sapiens 94-98 31559456-6 2019 RESULTS: HO-1 expression in HepG2 cells was increased by the treatment of cisplatin (CDDP) and pirarubicin (THP) dose-dependently. Cisplatin 74-83 heme oxygenase 1 Homo sapiens 9-13 31559456-6 2019 RESULTS: HO-1 expression in HepG2 cells was increased by the treatment of cisplatin (CDDP) and pirarubicin (THP) dose-dependently. Cisplatin 85-89 heme oxygenase 1 Homo sapiens 9-13 31404677-8 2019 RESULTS: mRNAs of PAI-1, HO-1, and p21 were upregulated after photoirradiation of HEK293, which was suppressed by N-acetyl-L-cysteine, a reactive oxygen species (ROS) scavenger. Reactive Oxygen Species 137-160 heme oxygenase 1 Homo sapiens 25-29 31404677-8 2019 RESULTS: mRNAs of PAI-1, HO-1, and p21 were upregulated after photoirradiation of HEK293, which was suppressed by N-acetyl-L-cysteine, a reactive oxygen species (ROS) scavenger. Reactive Oxygen Species 162-165 heme oxygenase 1 Homo sapiens 25-29 31559456-11 2019 CONCLUSION: The cytotoxic effects of CDDP and THP were both enhanced under HO-1 knockdown conditions as well as under conditions that inhibit the activation pathway of HO-1 by EGFR inhibitors. pirarubicin 46-49 heme oxygenase 1 Homo sapiens 168-172 31404677-11 2019 CONCLUSION: ALA-PDD or ALA-PDT can result in rapid ROS-induced cell death and may decrease long-term recurrence rates through several pathways including the HO-1/p21 pathway. 5-amino levulinic acid 12-15 heme oxygenase 1 Homo sapiens 157-161 31404677-11 2019 CONCLUSION: ALA-PDD or ALA-PDT can result in rapid ROS-induced cell death and may decrease long-term recurrence rates through several pathways including the HO-1/p21 pathway. 5-amino levulinic acid 23-26 heme oxygenase 1 Homo sapiens 157-161 31559456-6 2019 RESULTS: HO-1 expression in HepG2 cells was increased by the treatment of cisplatin (CDDP) and pirarubicin (THP) dose-dependently. pirarubicin 95-106 heme oxygenase 1 Homo sapiens 9-13 31559456-6 2019 RESULTS: HO-1 expression in HepG2 cells was increased by the treatment of cisplatin (CDDP) and pirarubicin (THP) dose-dependently. pirarubicin 108-111 heme oxygenase 1 Homo sapiens 9-13 31559456-7 2019 In HO-1 knockdown HepG2 cells, the HO-1 was not expressed and the percentage of trypan blue-positive cells (dead cells) was significantly increased after treatment of CDDP and THP. Trypan Blue 80-91 heme oxygenase 1 Homo sapiens 3-7 31404677-11 2019 CONCLUSION: ALA-PDD or ALA-PDT can result in rapid ROS-induced cell death and may decrease long-term recurrence rates through several pathways including the HO-1/p21 pathway. Reactive Oxygen Species 51-54 heme oxygenase 1 Homo sapiens 157-161 31319135-5 2019 Reduced expression of NADPH dehydrogenase quinone 1 (NQO-1) and heme oxygenase 1 (HO-1) genes, which are well-characterized downstream targets of Nrf2-mediated transactivation, was also confirmed by using ALA and another Nrf2 activator, marliolide. Thioctic Acid 205-208 heme oxygenase 1 Homo sapiens 64-80 31559456-7 2019 In HO-1 knockdown HepG2 cells, the HO-1 was not expressed and the percentage of trypan blue-positive cells (dead cells) was significantly increased after treatment of CDDP and THP. Cisplatin 167-171 heme oxygenase 1 Homo sapiens 3-7 31559456-7 2019 In HO-1 knockdown HepG2 cells, the HO-1 was not expressed and the percentage of trypan blue-positive cells (dead cells) was significantly increased after treatment of CDDP and THP. pirarubicin 176-179 heme oxygenase 1 Homo sapiens 3-7 31559456-10 2019 In human hepatoblastoma specimens, 4 of the 5 patients (80%) showed HO-1 expression changed much stronger in the viable tumor cells after CITA therapy. cita 138-142 heme oxygenase 1 Homo sapiens 68-72 31559456-11 2019 CONCLUSION: The cytotoxic effects of CDDP and THP were both enhanced under HO-1 knockdown conditions as well as under conditions that inhibit the activation pathway of HO-1 by EGFR inhibitors. Cisplatin 37-41 heme oxygenase 1 Homo sapiens 75-79 31559456-11 2019 CONCLUSION: The cytotoxic effects of CDDP and THP were both enhanced under HO-1 knockdown conditions as well as under conditions that inhibit the activation pathway of HO-1 by EGFR inhibitors. Cisplatin 37-41 heme oxygenase 1 Homo sapiens 168-172 31319135-5 2019 Reduced expression of NADPH dehydrogenase quinone 1 (NQO-1) and heme oxygenase 1 (HO-1) genes, which are well-characterized downstream targets of Nrf2-mediated transactivation, was also confirmed by using ALA and another Nrf2 activator, marliolide. Thioctic Acid 205-208 heme oxygenase 1 Homo sapiens 82-86 31319135-5 2019 Reduced expression of NADPH dehydrogenase quinone 1 (NQO-1) and heme oxygenase 1 (HO-1) genes, which are well-characterized downstream targets of Nrf2-mediated transactivation, was also confirmed by using ALA and another Nrf2 activator, marliolide. marliolide 237-247 heme oxygenase 1 Homo sapiens 64-80 31319135-5 2019 Reduced expression of NADPH dehydrogenase quinone 1 (NQO-1) and heme oxygenase 1 (HO-1) genes, which are well-characterized downstream targets of Nrf2-mediated transactivation, was also confirmed by using ALA and another Nrf2 activator, marliolide. marliolide 237-247 heme oxygenase 1 Homo sapiens 82-86 31654068-9 2019 Besides, scutellarin induced the mRNA expressions of heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1). scutellarin 9-20 heme oxygenase 1 Homo sapiens 53-69 31771519-20 2019 Bilirubin levels may become elevated as a result of heme oxygenase-1 activation, occurring in exercise-induced acute kidney injury in patients with RHUC; this phenomenon suggests renal ischemia-reperfusion injury. Bilirubin 0-9 heme oxygenase 1 Homo sapiens 52-68 31772153-11 2019 Heme Oxygenase 1 (HMOX1) was among the top upregulated genes, whose induction was ROS-dependent and HMOX1 depletion enhanced chaetocin mediated TRAIL sensitization. chaetocin 125-134 heme oxygenase 1 Homo sapiens 0-16 31772153-11 2019 Heme Oxygenase 1 (HMOX1) was among the top upregulated genes, whose induction was ROS-dependent and HMOX1 depletion enhanced chaetocin mediated TRAIL sensitization. chaetocin 125-134 heme oxygenase 1 Homo sapiens 18-23 31772153-11 2019 Heme Oxygenase 1 (HMOX1) was among the top upregulated genes, whose induction was ROS-dependent and HMOX1 depletion enhanced chaetocin mediated TRAIL sensitization. chaetocin 125-134 heme oxygenase 1 Homo sapiens 100-105 31767834-8 2019 At last we show that PGK1 expression is downregulated in human necrotic femoral head tissues of DEX-taking patients, correlating with HO1 depletion. Dexamethasone 96-99 heme oxygenase 1 Homo sapiens 134-137 31725784-5 2019 The mineral absorption pathway genes, HMOX1 and VDR are involved in iron metabolism and response to vitamin D, respectively. Iron 68-72 heme oxygenase 1 Homo sapiens 38-43 31885775-5 2019 Therefore, regulation of Nrf2 and HO-1 expression by the PI3K/AKT pathway plays an important role in the regulation of the antioxidant and antiapoptotic responses in DRG cells in a high-glucose culture model. Glucose 186-193 heme oxygenase 1 Homo sapiens 34-38 31819369-5 2019 The mRNA and protein expression levels of redox transcription factor Nrf2 and the downstream effector SOD-1, SOD-2, NQO-1 and HO-1 were assayed to explore the detailed mechanism by which ICA alleviates ECM degradation. icariin 187-190 heme oxygenase 1 Homo sapiens 126-130 31885801-8 2019 Furthermore, we report on a C-terminal truncation of HO-1, which is generally considered as a signal molecule. Carbon 28-29 heme oxygenase 1 Homo sapiens 53-57 31644887-0 2019 Quercetin, but not rutin, attenuated hydrogen peroxide-induced cell damage via heme oxygenase-1 induction in endothelial cells. Quercetin 0-9 heme oxygenase 1 Homo sapiens 79-95 31644887-0 2019 Quercetin, but not rutin, attenuated hydrogen peroxide-induced cell damage via heme oxygenase-1 induction in endothelial cells. Hydrogen Peroxide 37-54 heme oxygenase 1 Homo sapiens 79-95 31644887-5 2019 It was found that quercetin upregulated HMOX1 expression to protect endothelial cells against oxidative stress, and the protective effects of quercetin on H2O2-induced endothelial cell damage (such as loss of cell viability and reduction of nitric oxide) could be abolished by the specific small-interfering RNA against HMOX1 expression or HMOX1 activity inhibitor. Quercetin 18-27 heme oxygenase 1 Homo sapiens 40-45 31644887-5 2019 It was found that quercetin upregulated HMOX1 expression to protect endothelial cells against oxidative stress, and the protective effects of quercetin on H2O2-induced endothelial cell damage (such as loss of cell viability and reduction of nitric oxide) could be abolished by the specific small-interfering RNA against HMOX1 expression or HMOX1 activity inhibitor. Quercetin 18-27 heme oxygenase 1 Homo sapiens 320-325 31644887-5 2019 It was found that quercetin upregulated HMOX1 expression to protect endothelial cells against oxidative stress, and the protective effects of quercetin on H2O2-induced endothelial cell damage (such as loss of cell viability and reduction of nitric oxide) could be abolished by the specific small-interfering RNA against HMOX1 expression or HMOX1 activity inhibitor. Quercetin 18-27 heme oxygenase 1 Homo sapiens 320-325 31815138-0 2019 Sirt5 Attenuates Cisplatin-Induced Acute Kidney Injury through Regulation of Nrf2/HO-1 and Bcl-2. Cisplatin 17-26 heme oxygenase 1 Homo sapiens 82-86 31815138-12 2019 The levels of Nrf2, HO-1, and Bcl-2 proteins in HK-2 cells were also decreased after CDDP treatment. Cisplatin 85-89 heme oxygenase 1 Homo sapiens 20-24 31815138-15 2019 Together, the results demonstrated that Sirt5 attenuated cisplatin-induced apoptosis and mitochondrial injury in human kidney HK-2 cells, possibly through the regulation of Nrf2/HO-1 and Bcl-2. Cisplatin 57-66 heme oxygenase 1 Homo sapiens 178-182 31644887-6 2019 In addition, the activation of ERK/Nrf2 signaling pathway was critical to the upregulation of HMOX1 induced by quercetin. Quercetin 111-120 heme oxygenase 1 Homo sapiens 94-99 31644887-8 2019 Therefore, quercetin could attenuate oxidative stress-induced endothelial cell damage at least partly through ERK/Nrf2/HMOX1 pathway. Quercetin 11-20 heme oxygenase 1 Homo sapiens 119-124 31725784-5 2019 The mineral absorption pathway genes, HMOX1 and VDR are involved in iron metabolism and response to vitamin D, respectively. Vitamin D 100-109 heme oxygenase 1 Homo sapiens 38-43 31279900-0 2019 O-GlcNAcylation-dependent upregulation of HO1 triggers ammonia-induced oxidative stress and senescence in hepatic encephalopathy. o-glcnacylation 0-15 heme oxygenase 1 Homo sapiens 42-45 31814878-0 2019 The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. eriodictyol 94-105 heme oxygenase 1 Homo sapiens 9-13 31711520-9 2019 The effects of decitabine on P15INK4B and TP53 in MDS cells after inhibiting HO-1 were detected by Western blotting. decitabine 15-25 heme oxygenase 1 Homo sapiens 77-81 31711520-16 2019 Using the E2F inhibitor HLM006474 and the EZH2 inhibitor JQEZ5, we showed that HO-1 could regulate EZH2 expression. HLM006474 24-33 heme oxygenase 1 Homo sapiens 79-83 31781345-0 2019 Methane Alleviates Acetaminophen-Induced Liver Injury by Inhibiting Inflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Apoptosis through the Nrf2/HO-1/NQO1 Signaling Pathway. Methane 0-7 heme oxygenase 1 Homo sapiens 161-165 31781345-0 2019 Methane Alleviates Acetaminophen-Induced Liver Injury by Inhibiting Inflammation, Oxidative Stress, Endoplasmic Reticulum Stress, and Apoptosis through the Nrf2/HO-1/NQO1 Signaling Pathway. Acetaminophen 19-32 heme oxygenase 1 Homo sapiens 161-165 31781345-10 2019 We found that a methane-rich medium decreased the levels of reactive oxygen species (DHE fluorescent staining), inhibited apoptosis (cell flow test), and regulated the Nrf2/HO-1/NQO1 signaling pathway. Methane 16-23 heme oxygenase 1 Homo sapiens 173-177 31781345-11 2019 Our data indicated that MRS prevented APAP-induced hepatic injury via anti-inflammatory, antioxidant, anti-ERS, and antiapoptotic properties involving the Nrf2/HO-1/NQO1 signaling pathway. Acetaminophen 38-42 heme oxygenase 1 Homo sapiens 160-164 31814878-0 2019 The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. Flavanones 34-44 heme oxygenase 1 Homo sapiens 9-13 31814878-0 2019 The Nrf2/HO-1 Axis as Targets for Flavanones: Neuroprotection by Pinocembrin, Naringenin, and Eriodictyol. pinocembrin 65-76 heme oxygenase 1 Homo sapiens 9-13 31462605-7 2019 Treatment with DDC for 24 hours increased the expression of heme oxygenase-1 (HO-1), and this was controlled by the activation of the Nrf2-ARE pathway. 2',3'-dihydroxy-4',6'-dimethoxychalcone 15-18 heme oxygenase 1 Homo sapiens 60-76 31462615-9 2019 Treatment with DDC for 24 h also increased the expression levels of heme oxygenase-1 (HO-1) in a concentration-dependent manner. 2',3'-dihydroxy-4',6'-dimethoxychalcone 15-18 heme oxygenase 1 Homo sapiens 68-84 31462615-9 2019 Treatment with DDC for 24 h also increased the expression levels of heme oxygenase-1 (HO-1) in a concentration-dependent manner. 2',3'-dihydroxy-4',6'-dimethoxychalcone 15-18 heme oxygenase 1 Homo sapiens 86-90 31462615-10 2019 Pretreatment with the HO-1 inhibitor followed by DDC treatment before UV-A irradiation for 24 h reduced ROS production and the cytoprotective effect. Reactive Oxygen Species 104-107 heme oxygenase 1 Homo sapiens 22-26 31272005-10 2019 N-acetylcysteine antagonized arsenite-mediated induction of HMOX1 expression as well as reduction of neuronal and oligodendrocyte differentiation in hNPC suggesting involvement of oxidative stress in arsenite DNT. Acetylcysteine 0-16 heme oxygenase 1 Homo sapiens 60-65 31272005-10 2019 N-acetylcysteine antagonized arsenite-mediated induction of HMOX1 expression as well as reduction of neuronal and oligodendrocyte differentiation in hNPC suggesting involvement of oxidative stress in arsenite DNT. arsenite 29-37 heme oxygenase 1 Homo sapiens 60-65 31577640-5 2019 Mechanistic studies have revealed superoxide dismutase, heme oxygenase-1 and nuclear factor erythroid 2-related factor 2 as emerging targets for the beneficial effects of curcumin on the vasculature. Curcumin 171-179 heme oxygenase 1 Homo sapiens 56-120 31279900-0 2019 O-GlcNAcylation-dependent upregulation of HO1 triggers ammonia-induced oxidative stress and senescence in hepatic encephalopathy. Ammonia 55-62 heme oxygenase 1 Homo sapiens 42-45 31279900-4 2019 Mechanisms and functional consequences of HO1 upregulation were studied in NH4Cl-exposed astrocytes in vitro by western blot, qPCR and super-resolution microscopy. Ammonium Chloride 75-80 heme oxygenase 1 Homo sapiens 42-45 31279900-5 2019 RESULTS: HO1 and the endoplasmic reticulum (ER) stress marker grp78 were upregulated, together with changes in the expression of multiple iron metabolism-related genes, in post-mortem brain samples from patients with liver cirrhosis and HE. Iron 138-142 heme oxygenase 1 Homo sapiens 9-12 31279900-7 2019 Upregulation of HO1 by NH4Cl triggered ER stress and was associated with elevated levels of free ferrous iron and expression changes in iron metabolism-related genes, which were largely abolished after knockdown or inhibition of GS, GFAT1/2, HO1 or iron chelation. ferrous iron 97-109 heme oxygenase 1 Homo sapiens 16-19 31279900-7 2019 Upregulation of HO1 by NH4Cl triggered ER stress and was associated with elevated levels of free ferrous iron and expression changes in iron metabolism-related genes, which were largely abolished after knockdown or inhibition of GS, GFAT1/2, HO1 or iron chelation. Iron 105-109 heme oxygenase 1 Homo sapiens 16-19 31279900-7 2019 Upregulation of HO1 by NH4Cl triggered ER stress and was associated with elevated levels of free ferrous iron and expression changes in iron metabolism-related genes, which were largely abolished after knockdown or inhibition of GS, GFAT1/2, HO1 or iron chelation. Iron 136-140 heme oxygenase 1 Homo sapiens 16-19 31279900-10 2019 CONCLUSION: The present study identified glucosamine synthesis-dependent protein O-GlcNAcylation as a novel mechanism in the pathogenesis of HE that triggers oxidative and ER stress, as well as senescence, through upregulation of HO1 and Nox4. Glucosamine 41-52 heme oxygenase 1 Homo sapiens 230-233 31770287-9 2019 CONCLUSION: Propofol-based TIVA attenuated inflammatory response (elevated IL-1RA and IL-10 levels), downregulated MMP-9 response, and increased HO-1 expression with improved recovery of graft function and better microcirculation compared with desflurane anesthesia in liver transplant recipients. Propofol 12-20 heme oxygenase 1 Homo sapiens 145-149 31415221-6 2019 We found that dermaprazole induced nuclear translocation of erythroid 2-related factor 2 (Nrf2) and significantly upregulated heme oxygenase 1 (HO1) gene and protein expression in a 3D human skin model. dermaprazole 14-26 heme oxygenase 1 Homo sapiens 126-142 31770287-0 2019 Propofol vs desflurane on the cytokine, matrix metalloproteinase-9, and heme oxygenase-1 response during living donor liver transplantation: A pilot study. Propofol 0-8 heme oxygenase 1 Homo sapiens 72-88 31076999-0 2019 Naringenin Inhibit the Hydrogen Peroxide-Induced SH-SY5Y Cells Injury Through Nrf2/HO-1 Pathway. naringenin 0-10 heme oxygenase 1 Homo sapiens 83-87 31076999-0 2019 Naringenin Inhibit the Hydrogen Peroxide-Induced SH-SY5Y Cells Injury Through Nrf2/HO-1 Pathway. Hydrogen Peroxide 23-40 heme oxygenase 1 Homo sapiens 83-87 31076999-8 2019 NGN activated both ERK and PI3 K/Akt, and treatments with the specific ERK inhibitor PD98059, the PI3 K inhibitor LY294002, and the specific Nrf2 shRNA suppressed the NGN-induced HO-1 expression. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 85-92 heme oxygenase 1 Homo sapiens 179-183 31076999-8 2019 NGN activated both ERK and PI3 K/Akt, and treatments with the specific ERK inhibitor PD98059, the PI3 K inhibitor LY294002, and the specific Nrf2 shRNA suppressed the NGN-induced HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 114-122 heme oxygenase 1 Homo sapiens 179-183 31076999-9 2019 The HO-1 inhibitor ZnPP abolished the neuroprotective effect of NGN against H2O2-induced neurotoxicity. zinc protoporphyrin 19-23 heme oxygenase 1 Homo sapiens 4-8 31076999-9 2019 The HO-1 inhibitor ZnPP abolished the neuroprotective effect of NGN against H2O2-induced neurotoxicity. naringenin 64-67 heme oxygenase 1 Homo sapiens 4-8 31076999-9 2019 The HO-1 inhibitor ZnPP abolished the neuroprotective effect of NGN against H2O2-induced neurotoxicity. Hydrogen Peroxide 76-80 heme oxygenase 1 Homo sapiens 4-8 31076999-10 2019 Taken together, the present study demonstrates that regulation of Nrf2/HO-1 pathway through activation of ERK and PI3 K/Akt, and the inhibition of mitochondria-dependent apoptosis together may render NGN protect SH-SY5Y cells from H2O2-induced neurotoxicity. Hydrogen Peroxide 231-235 heme oxygenase 1 Homo sapiens 71-75 31415221-6 2019 We found that dermaprazole induced nuclear translocation of erythroid 2-related factor 2 (Nrf2) and significantly upregulated heme oxygenase 1 (HO1) gene and protein expression in a 3D human skin model. dermaprazole 14-26 heme oxygenase 1 Homo sapiens 144-147 31472180-9 2019 Additionally, the LCA treatment effectively counteracted TNF-alpha-mediated downregulation of silent information regulator 1 (SIRT1), nuclear factor erythroid2-related factor 2 (Nrf2), and heme oxygenase-1, which are related to oxidative stress. Lithocholic Acid 18-21 heme oxygenase 1 Homo sapiens 189-205 31749701-9 2019 Mechanistically, we found that GAS protects HUVECs from TBHP-induced cellular apoptosis by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. gastrodin 31-34 heme oxygenase 1 Homo sapiens 157-173 31648725-4 2019 In our previous study, Xanthohumol (Xn), a prenylated flavonoid extracted for hops (Humulus lupulus L), was screened from 386 natural products to inhibit PRRSV proliferation and alleviate oxidative stress induced by PRRSV via the Nrf2-HMOX1 axis in Marc-145 cells. xanthohumol 23-34 heme oxygenase 1 Homo sapiens 235-240 31648725-4 2019 In our previous study, Xanthohumol (Xn), a prenylated flavonoid extracted for hops (Humulus lupulus L), was screened from 386 natural products to inhibit PRRSV proliferation and alleviate oxidative stress induced by PRRSV via the Nrf2-HMOX1 axis in Marc-145 cells. xanthohumol 36-38 heme oxygenase 1 Homo sapiens 235-240 31749701-9 2019 Mechanistically, we found that GAS protects HUVECs from TBHP-induced cellular apoptosis by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. gastrodin 31-34 heme oxygenase 1 Homo sapiens 175-179 31749701-9 2019 Mechanistically, we found that GAS protects HUVECs from TBHP-induced cellular apoptosis by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. tert-Butylhydroperoxide 56-60 heme oxygenase 1 Homo sapiens 157-173 31749701-9 2019 Mechanistically, we found that GAS protects HUVECs from TBHP-induced cellular apoptosis by activating the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. tert-Butylhydroperoxide 56-60 heme oxygenase 1 Homo sapiens 175-179 31656916-7 2019 Furthermore, resveratrol and curcumin decreased the content of m6A and decreased the enrichment of m6A on the transcripts of tight junction proteins and on heme oxygenase-1 in the intestine. resveratrol 13-24 heme oxygenase 1 Homo sapiens 156-172 31635102-12 2019 (4) Conclusions: Microglial HO-1 induction with endogenous CO production functions as a crucial signaling pathway in blood-induced inflammation, determining microglial MCP-1 production and the extent of neuronal cell death. Carbon Dioxide 59-61 heme oxygenase 1 Homo sapiens 28-32 31635294-5 2019 We found that AGR pretreatment strongly inhibits the production of nitric oxide (NO), cytokines, and the enzymes inducible nitric oxide synthase (iNOS), and cyclooxygenase (COX)-2, and effectively induces the activation of heme oxygenase (HO)-1 and its regulator, nuclear factor erythroid 2-related factor 2 (Nrf-2). alanylglycylarginine 14-17 heme oxygenase 1 Homo sapiens 223-244 31330223-5 2019 The results revealed that SA4503 treatment increased Nrf2 and HO-1 expression significantly. SA 4503 26-32 heme oxygenase 1 Homo sapiens 62-66 31772710-0 2019 Melatonin Reduces Androgen Production and Upregulates Heme Oxygenase-1 Expression in Granulosa Cells from PCOS Patients with Hypoestrogenia and Hyperandrogenia. Melatonin 0-9 heme oxygenase 1 Homo sapiens 54-70 31656916-7 2019 Furthermore, resveratrol and curcumin decreased the content of m6A and decreased the enrichment of m6A on the transcripts of tight junction proteins and on heme oxygenase-1 in the intestine. Curcumin 29-37 heme oxygenase 1 Homo sapiens 156-172 31597407-7 2019 Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. Glutamic Acid 131-140 heme oxygenase 1 Homo sapiens 313-329 31597407-7 2019 Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. Glutamic Acid 131-140 heme oxygenase 1 Homo sapiens 331-335 31597407-7 2019 Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. Cystine 123-130 heme oxygenase 1 Homo sapiens 313-329 31597407-7 2019 Moreover, most of the enzymes and proteins that cascade or interact in the pathway of ferroptosis such as a subunit of the cystine/glutamate transporter xc- (SLC7A11), glutathione peroxidase 4 (GPX4), and the glutamate-cysteine ligase (GCLC) iron metabolism genes transferrin receptor 1 (TfR1) ferroportin, (Fpn) heme oxygenase 1 (HO-1) and ferritin are regulated by the antioxidant response element of the transcription factor, Nrf2. Cystine 123-130 heme oxygenase 1 Homo sapiens 331-335 31571584-6 2019 Complete heme tolerance requires a fully-operational heme degradation pathway as haplo insufficiency of HMOX1 combined with SLC48A1 inactivation causes perinatal lethality demonstrating synthetic lethal interactions between heme transport and degradation. Heme 9-13 heme oxygenase 1 Homo sapiens 104-109 31036877-5 2019 The knockdown of intracellular PRDX6 significantly enhances LOOH and ferroptotic cell death triggered by ferroptosis inducers (Erastin and RSL-3), which is correlated with the transcriptional activation of heme oxygenase-1. Lipid Peroxides 60-64 heme oxygenase 1 Homo sapiens 206-222 31036877-6 2019 Moreover, overexpression of heme oxygenase-1 enhances both Erastin- and RSL-3-triggered LOOH, suggesting that heme oxygenase-1 mediates PRDX6 silencing-enhanced ferroptosis. Lipid Peroxides 88-92 heme oxygenase 1 Homo sapiens 28-44 31036877-6 2019 Moreover, overexpression of heme oxygenase-1 enhances both Erastin- and RSL-3-triggered LOOH, suggesting that heme oxygenase-1 mediates PRDX6 silencing-enhanced ferroptosis. Lipid Peroxides 88-92 heme oxygenase 1 Homo sapiens 110-126 31243599-6 2019 Gene expression analysis revealed PL caused a > 20-fold induction of heme oxygenase-1 (HO-1), which we hypothesized was a survival mechanism for PDAC cells under enhanced oxidative stress. piperlonguminine 34-36 heme oxygenase 1 Homo sapiens 69-85 31243599-6 2019 Gene expression analysis revealed PL caused a > 20-fold induction of heme oxygenase-1 (HO-1), which we hypothesized was a survival mechanism for PDAC cells under enhanced oxidative stress. piperlonguminine 34-36 heme oxygenase 1 Homo sapiens 87-91 31243599-7 2019 HO-1 knockout resulted in enhanced PL-induced PDAC cell death under hypoxic conditions. piperlonguminine 35-37 heme oxygenase 1 Homo sapiens 0-4 31243599-8 2019 Similarly, high concentrations of the HO-1 inhibitor, ZnPP (10 microM), sensitized PDAC cells to PL; however, lower concentrations ZnPP (10 nM) and high or low concentrations of SnPP both protected PDAC cells from PL-induced cell death. zinc protoporphyrin 54-58 heme oxygenase 1 Homo sapiens 38-42 31243599-8 2019 Similarly, high concentrations of the HO-1 inhibitor, ZnPP (10 microM), sensitized PDAC cells to PL; however, lower concentrations ZnPP (10 nM) and high or low concentrations of SnPP both protected PDAC cells from PL-induced cell death. piperlonguminine 97-99 heme oxygenase 1 Homo sapiens 38-42 31243599-9 2019 Interestingly, the JNK inhibitor significantly blocked PL-induced PDAC cell death, Nrf-2 nuclear translocation, and HMOX-1 mRNA expression. piperlonguminine 55-57 heme oxygenase 1 Homo sapiens 116-122 31243599-10 2019 Collectively, the results demonstrate JNK signaling contributes to PL-induced PDAC cell death, and at the same time, activates Nrf-2 transcription of HMOX-1 as a compensatory survival mechanism. piperlonguminine 67-69 heme oxygenase 1 Homo sapiens 150-156 31188648-11 2019 Cell immunofluorescence staining indicated that the fluorescence staining intensity of HO-1 in the NBP group was significantly higher than that in the control group. 3-n-butylphthalide 99-102 heme oxygenase 1 Homo sapiens 87-91 31571584-6 2019 Complete heme tolerance requires a fully-operational heme degradation pathway as haplo insufficiency of HMOX1 combined with SLC48A1 inactivation causes perinatal lethality demonstrating synthetic lethal interactions between heme transport and degradation. Heme 53-57 heme oxygenase 1 Homo sapiens 104-109 31571584-6 2019 Complete heme tolerance requires a fully-operational heme degradation pathway as haplo insufficiency of HMOX1 combined with SLC48A1 inactivation causes perinatal lethality demonstrating synthetic lethal interactions between heme transport and degradation. Heme 53-57 heme oxygenase 1 Homo sapiens 104-109 31422179-6 2019 Moreover, the mRNA levels of Keap1, Nrf2, Maf and HO-1 indicated that l-carnitine regulated Nrf2/Keap1 activation. Carnitine 70-81 heme oxygenase 1 Homo sapiens 50-54 31325727-0 2019 Activation of Nrf2/HO-1 signal with Myricetin for attenuating ECM degradation in human chondrocytes and ameliorating the murine osteoarthritis. myricetin 36-45 heme oxygenase 1 Homo sapiens 19-23 31351365-1 2019 Naringenin (NG), a flavanone extracted from various plants, has potent vasoprotective effects likely related to the induction of heme oxygenase-1 (HO-1). naringenin 0-10 heme oxygenase 1 Homo sapiens 129-145 31351365-1 2019 Naringenin (NG), a flavanone extracted from various plants, has potent vasoprotective effects likely related to the induction of heme oxygenase-1 (HO-1). naringenin 0-10 heme oxygenase 1 Homo sapiens 147-151 31325727-11 2019 To explore the potential mechanism, we found out that myricetin suppressed NF-kappaB signaling pathway through Nrf2/HO-1 axis in human chondrocytes. myricetin 54-63 heme oxygenase 1 Homo sapiens 116-120 31351365-1 2019 Naringenin (NG), a flavanone extracted from various plants, has potent vasoprotective effects likely related to the induction of heme oxygenase-1 (HO-1). naringenin 12-14 heme oxygenase 1 Homo sapiens 129-145 31351365-1 2019 Naringenin (NG), a flavanone extracted from various plants, has potent vasoprotective effects likely related to the induction of heme oxygenase-1 (HO-1). naringenin 12-14 heme oxygenase 1 Homo sapiens 147-151 31351365-2 2019 In the current study, we investigated mechanisms underlying the effect of NG on HO-1 expression and high glucose (HG)- or free fatty acids (FFA)-induced apoptosis in human umbilical vein endothelial cells (HUVECs). naringenin 74-76 heme oxygenase 1 Homo sapiens 80-84 31351365-3 2019 First, we found that HUVECs exposed to NG exhibited enhanced HO-1 expression in a concentration- and time-dependent manner. naringenin 39-41 heme oxygenase 1 Homo sapiens 61-65 31351365-5 2019 LY294002 (a PI3K inhibitor) and SP600125 (a JNK inhibitor) reduced NG-induced HO-1 expression, whereas BIRB796 (a p38 inhibitor) and PD98059 (an ERK inhibitor) had no effect. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 heme oxygenase 1 Homo sapiens 78-82 31351365-5 2019 LY294002 (a PI3K inhibitor) and SP600125 (a JNK inhibitor) reduced NG-induced HO-1 expression, whereas BIRB796 (a p38 inhibitor) and PD98059 (an ERK inhibitor) had no effect. pyrazolanthrone 32-40 heme oxygenase 1 Homo sapiens 78-82 31325727-14 2019 CONCLUSION: Taken together, our data first demonstrated that myricetin possesses the therapeutic potential on OA through PI3K/Akt mediated Nrf2/HO-1 signaling pathway. myricetin 61-70 heme oxygenase 1 Homo sapiens 144-148 31356851-10 2019 Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. Hemin 86-91 heme oxygenase 1 Homo sapiens 14-30 30465316-5 2019 Stress and toxicity pathway analysis demonstrated that CAPE, in a dose- and time-related fashion, induced the expression of apoptotic and oxidative stress-response genes including growth arrest and DNA-damage inducible, alpha and gamma (GADD45A and GADD45G) and heme oxygenase-1. caffeic acid phenethyl ester 55-59 heme oxygenase 1 Homo sapiens 262-278 32587840-1 2019 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in the catalysis of heme conversion into biliverdin. Heme 76-80 heme oxygenase 1 Homo sapiens 0-16 32587840-1 2019 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in the catalysis of heme conversion into biliverdin. Heme 76-80 heme oxygenase 1 Homo sapiens 18-22 32587840-1 2019 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in the catalysis of heme conversion into biliverdin. Biliverdine 97-107 heme oxygenase 1 Homo sapiens 0-16 32587840-1 2019 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in the catalysis of heme conversion into biliverdin. Biliverdine 97-107 heme oxygenase 1 Homo sapiens 18-22 31264739-0 2019 Overexpression of heme oxygenase-1 in microenvironment mediates vincristine resistance of B-cell acute lymphoblastic leukemia by promoting vascular endothelial growth factor secretion. Vincristine 64-75 heme oxygenase 1 Homo sapiens 18-34 31264739-3 2019 Herein, overexpression of heme oxygenase-1 (HO-1) was found in the bone marrow stromal cells (BMSCs) from B-ALL patients developing resistance to vincristine (VCR), a chemotherapeutic agent. Vincristine 146-157 heme oxygenase 1 Homo sapiens 26-42 31264739-3 2019 Herein, overexpression of heme oxygenase-1 (HO-1) was found in the bone marrow stromal cells (BMSCs) from B-ALL patients developing resistance to vincristine (VCR), a chemotherapeutic agent. Vincristine 146-157 heme oxygenase 1 Homo sapiens 44-48 31356851-10 2019 Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. Hemin 96-101 heme oxygenase 1 Homo sapiens 14-30 31356851-10 2019 Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. N-methyl-4-aminoantipyrine 102-105 heme oxygenase 1 Homo sapiens 14-30 31356851-10 2019 Inhibition of heme oxygenase-1 or cytochrome P450 reductase increased the toxicity of hemin and hemin/MAA in undifferentiated, but only for hemin in differentiated HL60 cells. Hemin 96-101 heme oxygenase 1 Homo sapiens 14-30 31425676-2 2019 In a few instances e.g., neonatal jaundice, overproduction of HO-1 and increased HO activity results in elevated levels of bilirubin requiring clinical intervention with inhibitors of HO activity. Bilirubin 123-132 heme oxygenase 1 Homo sapiens 62-66 31425676-3 2019 In contrast HO-1 levels and HO activity are low in obesity and the HO system responds to mitigate the deleterious effects of oxidative stress through increased levels of bilirubin (anti-inflammatory) and CO (anti-apoptotic) and decreased levels of heme (pro-oxidant). Bilirubin 170-179 heme oxygenase 1 Homo sapiens 12-16 31319021-1 2019 Interest in the therapeutic effects of carbon monoxide (CO), a product of heme degradation catalyzed by the enzyme heme oxygenase-1 (HO-1), has led to the development of CO-releasing molecules (CO-RMs) for the controlled delivery of this gas in vivo. Carbon Monoxide 39-54 heme oxygenase 1 Homo sapiens 115-131 31067610-0 2019 Nrf2-Heme oxygenase-1 modulates autophagy and inhibits apoptosis triggered by elevated glucose levels in renal tubule cells. Glucose 87-94 heme oxygenase 1 Homo sapiens 5-21 31067610-10 2019 Conclusion: The activation of nuclear factor E2-related factor 2 (Nrf2)-HO-1 inhibited ROS expression and subsequently attenuated autophagy and cell apoptosis after HG injury was decreased. Reactive Oxygen Species 87-90 heme oxygenase 1 Homo sapiens 72-76 31067610-12 2019 Therefore, HO-1 activation can prevent ROS development and oxidative stress during HG injury, which considerably decreases autophagy and apoptosis. Reactive Oxygen Species 39-42 heme oxygenase 1 Homo sapiens 11-15 31547608-8 2019 Phenolic extracts derived from RB down-regulated the expression of four genes, ICAM1, CD39, CD73 and NOX4 and up-regulated the expression of another four genes, Nrf2, NQO1, HO1 and eNOS, indicating an antioxidant/ anti-inflammatory effect for RB against endothelial dysfunction. phenolic acid 0-8 heme oxygenase 1 Homo sapiens 173-176 31319021-1 2019 Interest in the therapeutic effects of carbon monoxide (CO), a product of heme degradation catalyzed by the enzyme heme oxygenase-1 (HO-1), has led to the development of CO-releasing molecules (CO-RMs) for the controlled delivery of this gas in vivo. Carbon Monoxide 39-54 heme oxygenase 1 Homo sapiens 133-137 31319021-1 2019 Interest in the therapeutic effects of carbon monoxide (CO), a product of heme degradation catalyzed by the enzyme heme oxygenase-1 (HO-1), has led to the development of CO-releasing molecules (CO-RMs) for the controlled delivery of this gas in vivo. Carbon Monoxide 56-58 heme oxygenase 1 Homo sapiens 115-131 31319021-1 2019 Interest in the therapeutic effects of carbon monoxide (CO), a product of heme degradation catalyzed by the enzyme heme oxygenase-1 (HO-1), has led to the development of CO-releasing molecules (CO-RMs) for the controlled delivery of this gas in vivo. Carbon Monoxide 56-58 heme oxygenase 1 Homo sapiens 133-137 31319021-2 2019 We recently proposed conjugating a cobalt-based CO-RM with various activators of nuclear factor erythroid 2-related factor 2 (Nrf2), the transcription factor that regulates HO-1 expression, in order to exploit the beneficial effects of exogenous and endogenous CO. Cobalt 35-41 heme oxygenase 1 Homo sapiens 173-177 31636810-0 2019 Lysosomal Destabilizing Drug Siramesine and the Dual Tyrosine Kinase Inhibitor Lapatinib Induce a Synergistic Ferroptosis through Reduced Heme Oxygenase-1 (HO-1) Levels. Lu 28-179 29-39 heme oxygenase 1 Homo sapiens 138-154 31533049-0 2019 Microanatomic Distribution of Myeloid Heme Oxygenase-1 Protects against Free Radical-Mediated Immunopathology in Human Tuberculosis. Free Radicals 72-84 heme oxygenase 1 Homo sapiens 38-54 31636810-0 2019 Lysosomal Destabilizing Drug Siramesine and the Dual Tyrosine Kinase Inhibitor Lapatinib Induce a Synergistic Ferroptosis through Reduced Heme Oxygenase-1 (HO-1) Levels. Lu 28-179 29-39 heme oxygenase 1 Homo sapiens 156-160 31636810-0 2019 Lysosomal Destabilizing Drug Siramesine and the Dual Tyrosine Kinase Inhibitor Lapatinib Induce a Synergistic Ferroptosis through Reduced Heme Oxygenase-1 (HO-1) Levels. Lapatinib 79-88 heme oxygenase 1 Homo sapiens 138-154 31636810-0 2019 Lysosomal Destabilizing Drug Siramesine and the Dual Tyrosine Kinase Inhibitor Lapatinib Induce a Synergistic Ferroptosis through Reduced Heme Oxygenase-1 (HO-1) Levels. Lapatinib 79-88 heme oxygenase 1 Homo sapiens 156-160 31636810-8 2019 Overexpression of HO-1 resulted in reduction of ROS and lipid peroxidation production and cell death. Reactive Oxygen Species 48-51 heme oxygenase 1 Homo sapiens 18-22 31636810-9 2019 Furthermore, knocking down of HO-1 combined with siramesine treatment resulted in increased cell death. Lu 28-179 49-59 heme oxygenase 1 Homo sapiens 30-34 31319070-9 2019 Furthermore, nicorandil (3 muM) enhanced nuclear factor erythroid 2-related factor 2 (Nrf2) translocation and heme oxygenase-1 (HO-1) expression. Nicorandil 13-23 heme oxygenase 1 Homo sapiens 110-126 31421070-6 2019 In fact, HO1, NADPH-cytochrome P450 reductase, and PCBP2 form a functional unit that integrates the catabolism of heme with the binding and transport of iron by PCBP2. Heme 114-118 heme oxygenase 1 Homo sapiens 9-12 31421070-6 2019 In fact, HO1, NADPH-cytochrome P450 reductase, and PCBP2 form a functional unit that integrates the catabolism of heme with the binding and transport of iron by PCBP2. Iron 153-157 heme oxygenase 1 Homo sapiens 9-12 31422075-6 2019 Bach1 silencing significantly enhanced sulforaphane-induced expression of HO-1 but had no effect on that of GCLC, GCLM, and NQO1 in young HBE cells. sulforaphane 39-51 heme oxygenase 1 Homo sapiens 74-78 31422075-7 2019 In contrast, Bach1 silencing enhanced sulforaphane-induced expression of GCLC, GCLM and HO-1 but had no effect on that of NQO-1 in older HBE cells. sulforaphane 38-50 heme oxygenase 1 Homo sapiens 88-92 31319070-9 2019 Furthermore, nicorandil (3 muM) enhanced nuclear factor erythroid 2-related factor 2 (Nrf2) translocation and heme oxygenase-1 (HO-1) expression. Nicorandil 13-23 heme oxygenase 1 Homo sapiens 128-132 31319070-11 2019 In summary, nicorandil may protect HUVECs from DOX-induced apoptosis, in part through ATF3-mediated Nrf2/HO-1 signaling pathways, which potentially protect the vessels from severe DOX toxicity. Nicorandil 12-22 heme oxygenase 1 Homo sapiens 105-109 31319070-11 2019 In summary, nicorandil may protect HUVECs from DOX-induced apoptosis, in part through ATF3-mediated Nrf2/HO-1 signaling pathways, which potentially protect the vessels from severe DOX toxicity. Doxorubicin 47-50 heme oxygenase 1 Homo sapiens 105-109 31506103-0 2019 Xanthohumol inhibits PRRSV proliferation and alleviates oxidative stress induced by PRRSV via the Nrf2-HMOX1 axis. xanthohumol 0-11 heme oxygenase 1 Homo sapiens 103-108 31506475-5 2019 DHA increased glutathione peroxidase activity (control, 3.2 +- 0.3 to 4.1 +- 0.2 nmol/min/ml/mug protein, p = 0.004; AAA, 2.3 +- 0.5 to 3.4 +- 0.5 nmol/min/ml/mug protein, p = 0.008) and heme oxygenase-1 mRNA expression (control, 1.5-fold increase, p < 0.001). Docosahexaenoic Acids 0-3 heme oxygenase 1 Homo sapiens 187-203 31487814-3 2019 Furthermore, Gln exerts potent antioxidant and anti-inflammatory effects in the circulation by inducing the expression of heme oxygenase-1, heat shock proteins, and glutathione. Glutamine 13-16 heme oxygenase 1 Homo sapiens 122-138 31564855-8 2019 More important, the genotypes, AG, GG of GSTP1 exon5 and L/M*S, L/L of HO-1 (GT)n associated with increased 8-iso-prostaglandin F (2 alpha) (8-iso-PGF2) and malondialdehyde (MDA) concentration and decreased catalase (CAT) activity. 8-isoprostaglandin A2 108-129 heme oxygenase 1 Homo sapiens 71-75 31491954-16 2019 In conclusion, cyanate could induce oxidative stress damage and lipid deposition by inhibiting Nrf2/HO-1 pathway, which was rescued by inhibitor of Nrf2. Cyanates 15-22 heme oxygenase 1 Homo sapiens 100-104 33448816-6 2019 HUVECs exposed to Rb-CA gels exhibited enhanced migration and tubule formation ability, increased vascular endothelial growth factor secretion, and improved activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2)/heme-oxygenase-1 (HO-1) signaling pathway. Rubidium 18-20 heme oxygenase 1 Homo sapiens 226-242 33448816-6 2019 HUVECs exposed to Rb-CA gels exhibited enhanced migration and tubule formation ability, increased vascular endothelial growth factor secretion, and improved activation of the nuclear factor (erythroid-derived 2)-like 2 (NRF2)/heme-oxygenase-1 (HO-1) signaling pathway. Rubidium 18-20 heme oxygenase 1 Homo sapiens 244-248 31564855-8 2019 More important, the genotypes, AG, GG of GSTP1 exon5 and L/M*S, L/L of HO-1 (GT)n associated with increased 8-iso-prostaglandin F (2 alpha) (8-iso-PGF2) and malondialdehyde (MDA) concentration and decreased catalase (CAT) activity. 8-epi-prostaglandin F2alpha 141-151 heme oxygenase 1 Homo sapiens 71-75 31564855-8 2019 More important, the genotypes, AG, GG of GSTP1 exon5 and L/M*S, L/L of HO-1 (GT)n associated with increased 8-iso-prostaglandin F (2 alpha) (8-iso-PGF2) and malondialdehyde (MDA) concentration and decreased catalase (CAT) activity. Malondialdehyde 157-172 heme oxygenase 1 Homo sapiens 71-75 31564855-8 2019 More important, the genotypes, AG, GG of GSTP1 exon5 and L/M*S, L/L of HO-1 (GT)n associated with increased 8-iso-prostaglandin F (2 alpha) (8-iso-PGF2) and malondialdehyde (MDA) concentration and decreased catalase (CAT) activity. Malondialdehyde 174-177 heme oxygenase 1 Homo sapiens 71-75 30861304-4 2019 Si50 (200 microg/mL) induced a time-dependent ROS formation and a postponed increase in expression of haem oxygenase (HO-1) mRNA and protein. si50 0-4 heme oxygenase 1 Homo sapiens 118-122 31075320-7 2019 Upon application of low doses of ALA-PDT to photoaging HDFs, Nrf2 was translocated to the nucleus; in addition, the expression of Nrf2, transforming growth factor-beta1 (TGF-beta1), type I and III collagen (COL1 and COL3), heme oxygenase 1 (HO-1), and p-ERK was increased, while the expression of matrix metalloproteinase 9 (MMP-9) was decreased. Aminolevulinic Acid 33-36 heme oxygenase 1 Homo sapiens 223-239 31146961-6 2019 On the protein-level, both cell types showed similar effects: At 5 mM HEMA, heme oxygenase-1 was induced 5.1-fold in OKF6/TERT2 cells and 4.1-fold in HGFs. hydroxyethyl methacrylate 70-74 heme oxygenase 1 Homo sapiens 76-92 31957703-0 2019 Dexmedetomidine alleviates hepatic injury via the inhibition of oxidative stress and activation of the Nrf2/HO-1 signaling pathway. Dexmedetomidine 0-15 heme oxygenase 1 Homo sapiens 108-112 31957703-9 2019 Moreover, Nrf2, HO-1, and Bcl-2 expression was upregulated, whereas, in contrast, transcripts for Bax, caspase3, and caspase9 were downregulated following Dex treatment under OGD/R. Dexmedetomidine 155-158 heme oxygenase 1 Homo sapiens 16-20 31957703-11 2019 Dex protects WRL-68 cells against OGD/R-induced injury by inhibiting inflammation, oxidative stress, and cell apoptosis via the activation of Nrf2/HO-1 signaling pathway, suggesting that Dex may be a potential protector against hepatic injury. Dexmedetomidine 0-3 heme oxygenase 1 Homo sapiens 147-151 31957703-11 2019 Dex protects WRL-68 cells against OGD/R-induced injury by inhibiting inflammation, oxidative stress, and cell apoptosis via the activation of Nrf2/HO-1 signaling pathway, suggesting that Dex may be a potential protector against hepatic injury. Dexmedetomidine 187-190 heme oxygenase 1 Homo sapiens 147-151 31226417-0 2019 Entinostat combined with Fludarabine synergistically enhances the induction of apoptosis in TP53 mutated CLL cells via the HDAC1/HO-1 pathway. fludarabine 25-36 heme oxygenase 1 Homo sapiens 129-133 31037501-0 2019 MiR-153 regulates cardiomyocyte apoptosis by targeting Nrf2/HO-1 signaling. mir-153 0-7 heme oxygenase 1 Homo sapiens 60-64 31037501-8 2019 Nrf2/ heme oxygenase-1 (HO-1) signaling plays a critical role in miR-153 regulated OGD/R-induced cardiomyocyte apoptosis. mir-153 65-72 heme oxygenase 1 Homo sapiens 6-22 31037501-8 2019 Nrf2/ heme oxygenase-1 (HO-1) signaling plays a critical role in miR-153 regulated OGD/R-induced cardiomyocyte apoptosis. mir-153 65-72 heme oxygenase 1 Homo sapiens 24-28 31112013-0 2019 Di (2-ethylhexyl) phthalate induces cytotoxicity in HEK-293 cell line, implication of the Nrf-2/HO-1 antioxidant pathway. Diethylhexyl Phthalate 0-27 heme oxygenase 1 Homo sapiens 96-100 31273911-8 2019 However, the process of HO-1 metabolism can be toxic owing to iron overload and the activation of succedent pathways, for example, the Fenton reaction and oxidative damage; the overall effect of HO-1 in SAH and ICH tends to be protective and harmful, respectively, given the different pathophysiological changes in these two types of haemorrhagic stroke. Iron 62-66 heme oxygenase 1 Homo sapiens 24-28 31075320-7 2019 Upon application of low doses of ALA-PDT to photoaging HDFs, Nrf2 was translocated to the nucleus; in addition, the expression of Nrf2, transforming growth factor-beta1 (TGF-beta1), type I and III collagen (COL1 and COL3), heme oxygenase 1 (HO-1), and p-ERK was increased, while the expression of matrix metalloproteinase 9 (MMP-9) was decreased. Aminolevulinic Acid 33-36 heme oxygenase 1 Homo sapiens 241-245 31456942-0 2019 SIRT5 Promotes Cisplatin Resistance in Ovarian Cancer by Suppressing DNA Damage in a ROS-Dependent Manner via Regulation of the Nrf2/HO-1 Pathway. Cisplatin 15-24 heme oxygenase 1 Homo sapiens 133-137 31173751-13 2019 Collectively, the results of this study suggest that Nrf2/HO-1 pathway has a protective role against CIN and support the clinical implication of Nrf2 activators, such as sulforaphane, in CIN particularly in diabetic patients. sulforaphane 170-182 heme oxygenase 1 Homo sapiens 58-62 31227482-0 2019 Isoflavone ME-344 Disrupts Redox Homeostasis and Mitochondrial Function by Targeting Heme Oxygenase 1. Isoflavones 0-10 heme oxygenase 1 Homo sapiens 85-101 31227482-3 2019 Drug-induced apoptosis was linked with enhanced levels of reactive oxygen species and this initiated a nuclear erythroid factor 2-like 2 signaling response, downstream of which, heme oxygenase 1 (HO-1) was also found to be time-dependently inhibited by ME-344. Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 178-194 31227482-3 2019 Drug-induced apoptosis was linked with enhanced levels of reactive oxygen species and this initiated a nuclear erythroid factor 2-like 2 signaling response, downstream of which, heme oxygenase 1 (HO-1) was also found to be time-dependently inhibited by ME-344. Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 196-200 31555758-5 2019 This review summarizes current knowledge of molecular mechanisms underlying renoprotective roles of EGCG in CKD based on available preclinical evidence (from both in vitro and in vivo animal studies), particularly its antioxidant property through preservation of mitochondrial function and activation of Nrf2 (nuclear factor erythroid 2-related factor 2)/HO-1 (heme oxygenase-1) signaling, anti-inflammatory activity, and protective effect against epithelial mesenchymal transition. epigallocatechin gallate 100-104 heme oxygenase 1 Homo sapiens 355-377 31181187-7 2019 Importantly, pelargonidin decreased the DNA methylation in the Nrf2 promoter region of JB6 P+ cells and increased Nrf2 downstream target genes expression, such as NAD(P)H/quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), involved in cellular protection. pelargonidin 13-25 heme oxygenase 1 Homo sapiens 207-223 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Heme 52-56 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Heme 52-56 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Heme 52-56 heme oxygenase 1 Homo sapiens 24-29 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 62-77 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 62-77 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 62-77 heme oxygenase 1 Homo sapiens 24-29 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 79-81 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 79-81 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Carbon Monoxide 79-81 heme oxygenase 1 Homo sapiens 24-29 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 84-96 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 84-96 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 84-96 heme oxygenase 1 Homo sapiens 24-29 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 98-102 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 98-102 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. ammonium ferrous sulfate 98-102 heme oxygenase 1 Homo sapiens 24-29 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Biliverdine 108-118 heme oxygenase 1 Homo sapiens 0-16 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Biliverdine 108-118 heme oxygenase 1 Homo sapiens 18-22 31276659-1 2019 Heme oxygenase-1 (HO-1, HMOX1) degrades pro-oxidant heme into carbon monoxide (CO), ferrous ions (Fe2+) and biliverdin. Biliverdine 108-118 heme oxygenase 1 Homo sapiens 24-29 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Cisplatin 51-60 heme oxygenase 1 Homo sapiens 259-275 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Cisplatin 51-60 heme oxygenase 1 Homo sapiens 277-281 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Cisplatin 105-114 heme oxygenase 1 Homo sapiens 259-275 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Cisplatin 105-114 heme oxygenase 1 Homo sapiens 277-281 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Reactive Oxygen Species 164-167 heme oxygenase 1 Homo sapiens 259-275 31456942-6 2019 Mechanistically, we show that SIRT5 contributes to cisplatin resistance in ovarian cancer by suppressing cisplatin-induced DNA damage in a reactive oxygen species (ROS)-dependent manner via regulation of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase 1 (HO-1) pathway. Reactive Oxygen Species 164-167 heme oxygenase 1 Homo sapiens 277-281 31467925-0 2019 Nrf2-Heme Oxygenase-1 Attenuates High-Glucose-Induced Epithelial-to-Mesenchymal Transition of Renal Tubule Cells by Inhibiting ROS-Mediated PI3K/Akt/GSK-3beta Signaling. Reactive Oxygen Species 127-130 heme oxygenase 1 Homo sapiens 5-21 31474865-5 2019 The present study showed that tyrosol, a phenylethanoid compound, suppresses accumulation of intracellular reactive oxygen species (ROS) caused by hyperglycemia, most plausibly by promoting heme oxygenase-1 (HO-1) expression in skeletal muscle cells. 4-hydroxyphenylethanol 30-37 heme oxygenase 1 Homo sapiens 190-206 31467925-6 2019 Treatment of HK2 cells with SFN caused HG-induced attenuation in EMT markers with activated Nrf2-HO-1. sulforaphane 28-31 heme oxygenase 1 Homo sapiens 97-101 31467925-9 2019 Conclusion: The present study suggests that Nrf2-HO-1 signaling has an inhibitory role in the regulation of EMT through the modulation of ROS-mediated PI3K/Akt/GSK-3beta activity, highlighting Nrf2-HO-1 and GSK-3beta as potential therapeutic targets in diabetic nephropathy. Reactive Oxygen Species 138-141 heme oxygenase 1 Homo sapiens 49-53 31467925-9 2019 Conclusion: The present study suggests that Nrf2-HO-1 signaling has an inhibitory role in the regulation of EMT through the modulation of ROS-mediated PI3K/Akt/GSK-3beta activity, highlighting Nrf2-HO-1 and GSK-3beta as potential therapeutic targets in diabetic nephropathy. Reactive Oxygen Species 138-141 heme oxygenase 1 Homo sapiens 198-202 31073781-9 2019 HO/HMOX/HSP32, expression was notably increased, which suggested the accumulation of reactive oxygen species. Reactive Oxygen Species 85-108 heme oxygenase 1 Homo sapiens 8-13 31141180-9 2019 These results demonstrated that 5-ALA/SFC treatment ameliorated binge alcohol exposure liver injury in a rat model of HIV-infected patients by reducing macrophage activation and expression of inflammatory cytokines/chemokines, and by inducing HO-1, HO-2, and Sirt1 expression. 5-amino levulinic acid 32-37 heme oxygenase 1 Homo sapiens 243-247 31141180-9 2019 These results demonstrated that 5-ALA/SFC treatment ameliorated binge alcohol exposure liver injury in a rat model of HIV-infected patients by reducing macrophage activation and expression of inflammatory cytokines/chemokines, and by inducing HO-1, HO-2, and Sirt1 expression. SFC 38-41 heme oxygenase 1 Homo sapiens 243-247 31030250-9 2019 Patients harboring mutant HMOX-1 had a less frequent vaso-occlusive crisis (VOC)/lifetime, less VOC in the last year, less incidence of stroke, less frequency of hospitalization, and responded more frequently to hydroxyurea with statistically significant differences (p = 0.028, 0.007, 0.046, 0.007, and 0.011 respectively). Hydroxyurea 212-223 heme oxygenase 1 Homo sapiens 26-32 31387260-7 2019 The activity of HO-1 was inhibited using Tin Mesoporphyrin IX (SnMP). tin mesoporphyrin ix 41-61 heme oxygenase 1 Homo sapiens 16-20 31387260-7 2019 The activity of HO-1 was inhibited using Tin Mesoporphyrin IX (SnMP). tin mesoporphyrin 63-67 heme oxygenase 1 Homo sapiens 16-20 30871975-5 2019 In this study we found that heme oxygenase-1, which could induce decay-accelerating factor (DAF) and inhibit the membrane-attack complex, was significantly decreased in patients with TA-TMA. ta-tma 183-189 heme oxygenase 1 Homo sapiens 28-44 31250343-13 2019 Moreover, the astragalin-induced the upregulation of HO-1 in human lung epithelial cells was inhibited when nuclear factor erythroid-2-related factor 2 (Nrf2) was silenced by small interfering RNA. astragalin 14-24 heme oxygenase 1 Homo sapiens 53-57 31250343-14 2019 Astragalin reduces LPS-induced ALI via activation of Nrf2/HO-1 pathway. astragalin 0-10 heme oxygenase 1 Homo sapiens 58-62 31153982-4 2019 Simultaneously, hesperetin activated the Nrf2 signaling pathway and regulated its downstream genes, including NQO1 and HO-1. hesperetin 16-26 heme oxygenase 1 Homo sapiens 119-123 31059786-0 2019 Lipoxin A4 restores oxidative stress-induced vascular endothelial cell injury and thrombosis-related factor expression by its receptor-mediated activation of Nrf2-HO-1 axis. lipoxin A4 0-10 heme oxygenase 1 Homo sapiens 163-167 31059786-9 2019 Mechanism analysis substantiated that LXA4 further augmented activation of the Nrf2-HO-1 pathway. lipoxin A4 38-42 heme oxygenase 1 Homo sapiens 84-88 31059786-10 2019 Nevertheless, blocking this signaling via si-Nrf2 transfection or HO-1 antagonist ZnPP both reversed LXA4-mediated effects against oxidative stress injury and thrombotic potential. Silicon 28-30 heme oxygenase 1 Homo sapiens 66-70 31059786-10 2019 Nevertheless, blocking this signaling via si-Nrf2 transfection or HO-1 antagonist ZnPP both reversed LXA4-mediated effects against oxidative stress injury and thrombotic potential. zinc protoporphyrin 82-86 heme oxygenase 1 Homo sapiens 66-70 31059786-11 2019 Cessation of the LXA4 receptor pathway by its inhibitor Boc2 not only counteracted LXA4-evoked activation of the Nrf2-HO-1, but also reversed LXA4-mediated anti-oxidative stress and thrombosis-related factor expression. boc2 56-60 heme oxygenase 1 Homo sapiens 118-122 31059786-12 2019 Accordingly, this study suggests that LXA4 may ameliorate vascular endothelial cell oxidative stress injury and subsequent thrombotic response via LXA4 receptor-dependent activation of the Nrf2-HO-1 signaling, implying a promising strategy for DVT and its complication. lipoxin A4 38-42 heme oxygenase 1 Homo sapiens 194-198 31202006-11 2019 Finally, only CY could lead to an increase in heme oxygenase 1 (HMOX1) expression. Cyclophosphamide 14-16 heme oxygenase 1 Homo sapiens 46-62 31172435-5 2019 Among the three types of SOA, m-xylene-derived SOA showed the strongest induction of the HMOX1 and IL8 genes, and transcriptional activity via the antioxidant response element (ARE). Xylenes 32-38 heme oxygenase 1 Homo sapiens 89-94 31202006-11 2019 Finally, only CY could lead to an increase in heme oxygenase 1 (HMOX1) expression. Cyclophosphamide 14-16 heme oxygenase 1 Homo sapiens 64-69 31389609-11 2019 High glucose notably lowered the protein expression levels of Nrf2, HO-1, and gamma-GCS in the cells (p<0.01). Glucose 5-12 heme oxygenase 1 Homo sapiens 68-72 30842612-3 2019 These alterations in HO-1 and NO markers were prevented by cotreatment with the polyphenol resveratrol, which also improved GSH levels. polyphenol resveratrol 80-102 heme oxygenase 1 Homo sapiens 21-25 30842612-3 2019 These alterations in HO-1 and NO markers were prevented by cotreatment with the polyphenol resveratrol, which also improved GSH levels. Glutathione 124-127 heme oxygenase 1 Homo sapiens 21-25 31143972-0 2019 Salsalate ameliorates the atherosclerotic response through HO-1- and SIRT1-mediated suppression of ER stress and inflammation. salicylsalicylic acid 0-9 heme oxygenase 1 Homo sapiens 59-63 31356549-0 2019 Curdione Ameliorated Doxorubicin-Induced Cardiotoxicity Through Suppressing Oxidative Stress and Activating Nrf2/HO-1 Pathway. curdione 0-8 heme oxygenase 1 Homo sapiens 113-117 31143972-12 2019 CONCLUSIONS: Salsalate ameliorates LPS-induced atherosclerotic reactions via HO-1 and SIRT1-dependent reduction of inflammation and ER stress. salicylsalicylic acid 13-22 heme oxygenase 1 Homo sapiens 77-81 31356549-0 2019 Curdione Ameliorated Doxorubicin-Induced Cardiotoxicity Through Suppressing Oxidative Stress and Activating Nrf2/HO-1 Pathway. Doxorubicin 21-32 heme oxygenase 1 Homo sapiens 113-117 31199053-2 2019 In the presence of stimulation by cobalt protoporphyrin (CoPP), an HO-1 inducer, apoptotic characteristics were observed, including DNA laddering, hypodiploid cells, and cleavages of caspase (Casp)-3 and poly(ADP) ribose polymerase (PARP) proteins in human colon carcinoma COLO205, HCT-15, LOVO and HT-29 cells in serum-free (SF) conditions with increased HO-1, but not heat shock protein 70 (HSP70) or HSP90. cobaltiprotoporphyrin 34-55 heme oxygenase 1 Homo sapiens 67-71 31199053-2 2019 In the presence of stimulation by cobalt protoporphyrin (CoPP), an HO-1 inducer, apoptotic characteristics were observed, including DNA laddering, hypodiploid cells, and cleavages of caspase (Casp)-3 and poly(ADP) ribose polymerase (PARP) proteins in human colon carcinoma COLO205, HCT-15, LOVO and HT-29 cells in serum-free (SF) conditions with increased HO-1, but not heat shock protein 70 (HSP70) or HSP90. cobaltiprotoporphyrin 34-55 heme oxygenase 1 Homo sapiens 356-360 31199053-2 2019 In the presence of stimulation by cobalt protoporphyrin (CoPP), an HO-1 inducer, apoptotic characteristics were observed, including DNA laddering, hypodiploid cells, and cleavages of caspase (Casp)-3 and poly(ADP) ribose polymerase (PARP) proteins in human colon carcinoma COLO205, HCT-15, LOVO and HT-29 cells in serum-free (SF) conditions with increased HO-1, but not heat shock protein 70 (HSP70) or HSP90. cobaltiprotoporphyrin 57-61 heme oxygenase 1 Homo sapiens 67-71 31199053-2 2019 In the presence of stimulation by cobalt protoporphyrin (CoPP), an HO-1 inducer, apoptotic characteristics were observed, including DNA laddering, hypodiploid cells, and cleavages of caspase (Casp)-3 and poly(ADP) ribose polymerase (PARP) proteins in human colon carcinoma COLO205, HCT-15, LOVO and HT-29 cells in serum-free (SF) conditions with increased HO-1, but not heat shock protein 70 (HSP70) or HSP90. cobaltiprotoporphyrin 57-61 heme oxygenase 1 Homo sapiens 356-360 31356549-9 2019 Further study indicated that curdione decreased DOX-induced phosphorylation of extracellular signal-regulated kinase1/2 (Erk1/2) and c-Jun-N-terminal kinase and activated nuclear factor-erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signal pathway. curdione 29-37 heme oxygenase 1 Homo sapiens 238-242 31356549-9 2019 Further study indicated that curdione decreased DOX-induced phosphorylation of extracellular signal-regulated kinase1/2 (Erk1/2) and c-Jun-N-terminal kinase and activated nuclear factor-erythroid 2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) signal pathway. Doxorubicin 48-51 heme oxygenase 1 Homo sapiens 238-242 30951941-6 2019 In addition, TBBPA promoted the expression of antioxidant genes related to Nrf2, such as quinone oxidoreductase 1 (NQO1), catalase (CAT), and heme oxygenase 1 (HO-1). tetrabromobisphenol A 13-18 heme oxygenase 1 Homo sapiens 142-158 30770549-0 2019 Curcumin prevents high glucose damage in retinal pigment epithelial cells through ERK1/2-mediated activation of the Nrf2/HO-1 pathway. Curcumin 0-8 heme oxygenase 1 Homo sapiens 121-125 30770549-5 2019 Further, curcumin was able to induce HO-1 expression via Nrf2 activation and counteracts the damage elicited by HG. Curcumin 9-17 heme oxygenase 1 Homo sapiens 37-41 30770549-6 2019 The present study demonstrated that curcumin provides protection against HG-induced damage in RPE cells through the activation of Nrf2/HO-1 signaling that involves the ERK pathway, suggesting that curcumin may have therapeutic value in the treatment of diabetic retinopathy. Curcumin 36-44 heme oxygenase 1 Homo sapiens 135-139 30770549-6 2019 The present study demonstrated that curcumin provides protection against HG-induced damage in RPE cells through the activation of Nrf2/HO-1 signaling that involves the ERK pathway, suggesting that curcumin may have therapeutic value in the treatment of diabetic retinopathy. Curcumin 197-205 heme oxygenase 1 Homo sapiens 135-139 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Heme 83-87 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Heme 83-87 heme oxygenase 1 Homo sapiens 18-22 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Iron 100-112 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Iron 100-112 heme oxygenase 1 Homo sapiens 18-22 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 114-129 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 114-129 heme oxygenase 1 Homo sapiens 18-22 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 131-133 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 131-133 heme oxygenase 1 Homo sapiens 18-22 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 18-22 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Bilirubin 187-196 heme oxygenase 1 Homo sapiens 0-16 31344980-1 2019 Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate ferrous iron, carbon monoxide (CO), and biliverdin, which is subsequently converted to bilirubin. Bilirubin 187-196 heme oxygenase 1 Homo sapiens 18-22 31344980-5 2019 In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. Heme 141-145 heme oxygenase 1 Homo sapiens 20-24 31344980-5 2019 In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. Biliverdine 179-189 heme oxygenase 1 Homo sapiens 20-24 31344980-5 2019 In atherosclerosis, HO-1 may play a protective role against the progression of atherosclerosis, mainly due to the degradation of pro-oxidant heme, the generation of anti-oxidants biliverdin and bilirubin and the production of vasodilator CO. Bilirubin 194-203 heme oxygenase 1 Homo sapiens 20-24 31214670-5 2019 Furthermore, DHA promoted the nuclear translocation of nuclear factor E2-related factor 2 (Nrf2), wherein Nrf2 further mediated the expression of multiple antioxidant enzymes such as, heme oxygenase-1 (HO-1), NAD(P)H: quinone oxidoreductase-1 (NQO1) and catalase, which are closely related with redox homeostasis. Docosahexaenoic Acids 13-16 heme oxygenase 1 Homo sapiens 184-200 31396016-4 2019 Both conditions can be accompanied by decreased levels of heme oxygenase-1 (HMOX1), cytoprotective, heme-degrading enzyme. Heme 58-62 heme oxygenase 1 Homo sapiens 76-81 31396016-10 2019 On the other hand, the only one that induced heme oxygenase-1 expression was sulforaphane, a known activator of a HMOX1 inducer-NRF2. sulforaphane 77-89 heme oxygenase 1 Homo sapiens 45-61 31396016-10 2019 On the other hand, the only one that induced heme oxygenase-1 expression was sulforaphane, a known activator of a HMOX1 inducer-NRF2. sulforaphane 77-89 heme oxygenase 1 Homo sapiens 114-119 30776144-3 2019 The heme oxygenase 1, encoded by the HMOX1 gene and NADH:quinone oxidoreductase 1, encoded by the NQO1 gene are crucial mediators of cellular defense against ROS. Reactive Oxygen Species 158-161 heme oxygenase 1 Homo sapiens 4-20 30776144-3 2019 The heme oxygenase 1, encoded by the HMOX1 gene and NADH:quinone oxidoreductase 1, encoded by the NQO1 gene are crucial mediators of cellular defense against ROS. Reactive Oxygen Species 158-161 heme oxygenase 1 Homo sapiens 37-42 30776144-4 2019 The present study aimed to investigate the associations between HMOX1 and NQO1 polymorphisms and ATLI in Chinese anti-TB treatment population. Terbium 118-120 heme oxygenase 1 Homo sapiens 64-69 30776144-9 2019 WHAT IS NEW AND CONCLUSION: This is the first study to explore the relationship between HMOX1, NQO1 polymorphisms and ATLI in Chinese anti-TB treatment population. Terbium 139-141 heme oxygenase 1 Homo sapiens 88-93 31423266-0 2019 Stimulated upregulation of HO-1 is associated with inadequate response of gastric and ovarian cancer cell lines to hyperthermia and cisplatin treatment. Cisplatin 132-141 heme oxygenase 1 Homo sapiens 27-31 31423266-4 2019 In OVCAR-3 cells, cisplatin increased HO-1 mRNA expression by 3.73-fold under normothermia and 2.4-fold under hyperthermia; furthermore, these factors similarly increased HO-1 protein expression levels. Cisplatin 18-27 heme oxygenase 1 Homo sapiens 38-42 31423266-4 2019 In OVCAR-3 cells, cisplatin increased HO-1 mRNA expression by 3.73-fold under normothermia and 2.4-fold under hyperthermia; furthermore, these factors similarly increased HO-1 protein expression levels. Cisplatin 18-27 heme oxygenase 1 Homo sapiens 171-175 31423266-6 2019 HO-1-silencing under normothermia increased apoptotic rates in cisplatin-treated OVCAR-3 cells by 2.07-fold, and hyperthermia enhanced the effect by 3.09-fold. Cisplatin 63-72 heme oxygenase 1 Homo sapiens 0-4 31423266-13 2019 In tumors with highly inducible HO-1, prior silencing of this gene could improve the cellular response to hyperthermia and cisplatin. Cisplatin 123-132 heme oxygenase 1 Homo sapiens 32-36 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Heme 43-47 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Carbon Monoxide 70-72 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Iron 83-87 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Biliverdine 93-103 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Biliverdine 105-107 heme oxygenase 1 Homo sapiens 0-22 31396090-3 2019 Heme oxygenase (HO) -1 can catabolize free heme into carbon monoxide (CO), ferrous iron, and biliverdin (BV)/bilirubin (BR). Bilirubin 109-118 heme oxygenase 1 Homo sapiens 0-22 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. 5-amino levulinic acid 0-21 heme oxygenase 1 Homo sapiens 147-163 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. 5-amino levulinic acid 0-21 heme oxygenase 1 Homo sapiens 165-169 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. 5-amino levulinic acid 23-26 heme oxygenase 1 Homo sapiens 147-163 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. 5-amino levulinic acid 23-26 heme oxygenase 1 Homo sapiens 165-169 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. Ferromia 42-64 heme oxygenase 1 Homo sapiens 147-163 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. Ferromia 42-64 heme oxygenase 1 Homo sapiens 165-169 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. SFC 66-69 heme oxygenase 1 Homo sapiens 147-163 31332208-2 2019 5-Aminolevulinic acid (ALA) combined with sodium ferrous citrate (SFC) was reported to enhance haem production, leading to respiratory complex and haem oxygenase-1 (HO-1) upregulation. SFC 66-69 heme oxygenase 1 Homo sapiens 165-169 31332208-9 2019 Thus, our findings indicate that ALA/SFC is effective in elevating OXPHOS, HO-1 protein, and mtDNA copy number, resulting in an increase in OCR and ATP levels, which represents a promising therapeutic option for mitochondrial diseases. SFC 37-40 heme oxygenase 1 Homo sapiens 75-79 31379570-9 2019 Moreover, TTB potentially prolonged the activation state of the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway and maintained the protection against oxidative stress in OGD/R-induced astrocytes by inducing the nuclear translocation and up-regulation of Nrf2 along with the enhanced expression of the downstream target gene HO-1. Thenoyltrifluoroacetone 10-13 heme oxygenase 1 Homo sapiens 115-131 31257023-3 2019 Nrf2 accumulation in lung cancers causes the stabilization of Bach1 by inducing Ho1, the enzyme catabolizing heme. Heme 109-113 heme oxygenase 1 Homo sapiens 80-83 31360208-4 2019 We also noticed that BP restored Ox-LDL-stimulated HUVECs oxidative stress, by induced antioxidant gene expressions, including Heme oxygenase-1 and its upstream mediator, Nrf2, which were mediated by the activation of the phosphorylation of PI3K/Akt/mTOR. Benzo(a)pyrene 21-23 heme oxygenase 1 Homo sapiens 127-143 31360208-5 2019 Pretreatment with wortmannin, PI3K/AKT inhibitor, abolished BP induced Nrf2 nuclear translocation and HO-1 level. Wortmannin 18-28 heme oxygenase 1 Homo sapiens 102-106 31360208-6 2019 Our results demonstrated that BP protected HUVECs against oxidative damage partly via PI3K/Akt/mTOR-mediated Nrf/HO-1 pathway, which might be applied into preventing Ox-LDL mediated cardiovascular diseases. Benzo(a)pyrene 30-32 heme oxygenase 1 Homo sapiens 113-117 31277394-4 2019 Moreover, piceatannol treatment induced NF-E2-related factor 2 (Nrf2) signaling activation, which was evidenced by increased transcription of anti-oxidant genes, glutamate-cysteine ligase catalytic subunit (GCLc), SOD, and HO-1. 3,3',4,5'-tetrahydroxystilbene 10-21 heme oxygenase 1 Homo sapiens 223-227 31277394-5 2019 Knockdown of Nrf2 through targeted siRNA alleviated piceatannol-mediated HO-1 transcription, and significantly abolished piceatannol-mediated cytoprotection. 3,3',4,5'-tetrahydroxystilbene 52-63 heme oxygenase 1 Homo sapiens 73-77 31277394-6 2019 LY294002 (PI3K inhibitor) dramatically blocked piceatannol-mediated increasing of Nrf2 nuclear translocation, HO-1 expression, and cytoprotective activity, indicating the involvement of PI3K/Akt pathway in the cytoprotective effect of piceatannol. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 heme oxygenase 1 Homo sapiens 110-114 31277394-6 2019 LY294002 (PI3K inhibitor) dramatically blocked piceatannol-mediated increasing of Nrf2 nuclear translocation, HO-1 expression, and cytoprotective activity, indicating the involvement of PI3K/Akt pathway in the cytoprotective effect of piceatannol. 3,3',4,5'-tetrahydroxystilbene 47-58 heme oxygenase 1 Homo sapiens 110-114 31219431-0 2019 Activation of the Nrf2/HO-1 signaling pathway contributes to the protective effects of coptisine against oxidative stress-induced DNA damage and apoptosis in HaCaT keratinocytes. coptisine 87-96 heme oxygenase 1 Homo sapiens 23-27 31035147-9 2019 Furthermore, as the downstream components of Nrf2 pathway, catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPX) and heme oxygenase-1 (HO-1) were significantly synergistically induced by formaldehyde and acrolein mixtures. Formaldehyde 209-221 heme oxygenase 1 Homo sapiens 139-155 31035147-9 2019 Furthermore, as the downstream components of Nrf2 pathway, catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GPX) and heme oxygenase-1 (HO-1) were significantly synergistically induced by formaldehyde and acrolein mixtures. Acrolein 226-234 heme oxygenase 1 Homo sapiens 139-155 31219431-7 2019 Furthermore, the protective effects of coptisine against H2O2-induced cytotoxicity were abolished by ZnPP, indicating that coptisine protects keratinocytes against oxidative stress-induced injury through activation of the Nrf2/HO-1 signaling pathway. coptisine 39-48 heme oxygenase 1 Homo sapiens 227-231 31219431-7 2019 Furthermore, the protective effects of coptisine against H2O2-induced cytotoxicity were abolished by ZnPP, indicating that coptisine protects keratinocytes against oxidative stress-induced injury through activation of the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 57-61 heme oxygenase 1 Homo sapiens 227-231 31219431-4 2019 Interestingly, the expressions of nuclear factor-erythroid-2-related factor 2 (Nrf2) and its active form, phosphorylated Nrf2, were strikingly promoted by coptisine in the presence of H2O2, which was associated with a marked increase in the expression of heme oxygenase-1 (HO-1). coptisine 155-164 heme oxygenase 1 Homo sapiens 255-271 31219431-7 2019 Furthermore, the protective effects of coptisine against H2O2-induced cytotoxicity were abolished by ZnPP, indicating that coptisine protects keratinocytes against oxidative stress-induced injury through activation of the Nrf2/HO-1 signaling pathway. zinc protoporphyrin 101-105 heme oxygenase 1 Homo sapiens 227-231 31219431-4 2019 Interestingly, the expressions of nuclear factor-erythroid-2-related factor 2 (Nrf2) and its active form, phosphorylated Nrf2, were strikingly promoted by coptisine in the presence of H2O2, which was associated with a marked increase in the expression of heme oxygenase-1 (HO-1). coptisine 155-164 heme oxygenase 1 Homo sapiens 273-277 31219431-7 2019 Furthermore, the protective effects of coptisine against H2O2-induced cytotoxicity were abolished by ZnPP, indicating that coptisine protects keratinocytes against oxidative stress-induced injury through activation of the Nrf2/HO-1 signaling pathway. coptisine 123-132 heme oxygenase 1 Homo sapiens 227-231 31219431-4 2019 Interestingly, the expressions of nuclear factor-erythroid-2-related factor 2 (Nrf2) and its active form, phosphorylated Nrf2, were strikingly promoted by coptisine in the presence of H2O2, which was associated with a marked increase in the expression of heme oxygenase-1 (HO-1). Hydrogen Peroxide 184-188 heme oxygenase 1 Homo sapiens 255-271 31219431-4 2019 Interestingly, the expressions of nuclear factor-erythroid-2-related factor 2 (Nrf2) and its active form, phosphorylated Nrf2, were strikingly promoted by coptisine in the presence of H2O2, which was associated with a marked increase in the expression of heme oxygenase-1 (HO-1). Hydrogen Peroxide 184-188 heme oxygenase 1 Homo sapiens 273-277 31219431-5 2019 However, coptisine-induced HO-1 expression was completely abrogated by Nrf2-specific small interfering RNA (Nrf2-siRNA), which suggests that the increased expression of HO-1 by coptisine is Nrf2-dependent. coptisine 9-18 heme oxygenase 1 Homo sapiens 27-31 31219431-5 2019 However, coptisine-induced HO-1 expression was completely abrogated by Nrf2-specific small interfering RNA (Nrf2-siRNA), which suggests that the increased expression of HO-1 by coptisine is Nrf2-dependent. coptisine 9-18 heme oxygenase 1 Homo sapiens 169-173 31219431-5 2019 However, coptisine-induced HO-1 expression was completely abrogated by Nrf2-specific small interfering RNA (Nrf2-siRNA), which suggests that the increased expression of HO-1 by coptisine is Nrf2-dependent. coptisine 177-186 heme oxygenase 1 Homo sapiens 27-31 31219431-5 2019 However, coptisine-induced HO-1 expression was completely abrogated by Nrf2-specific small interfering RNA (Nrf2-siRNA), which suggests that the increased expression of HO-1 by coptisine is Nrf2-dependent. coptisine 177-186 heme oxygenase 1 Homo sapiens 169-173 31115551-0 2019 Glycyrrhizin protects human melanocytes from H2O2-induced oxidative damage via the Nrf2-dependent induction of HO-1. Glycyrrhizic Acid 0-12 heme oxygenase 1 Homo sapiens 111-115 31115551-0 2019 Glycyrrhizin protects human melanocytes from H2O2-induced oxidative damage via the Nrf2-dependent induction of HO-1. Hydrogen Peroxide 45-49 heme oxygenase 1 Homo sapiens 111-115 31115551-11 2019 The knockdown of Nrf2 by small interfering RNA or the inhibition of HO-1 by ZnPP reversed the protective effect of GR on melanocytes against H2O2-induced cytotoxicity and apoptosis. zinc protoporphyrin 76-80 heme oxygenase 1 Homo sapiens 68-72 31115551-11 2019 The knockdown of Nrf2 by small interfering RNA or the inhibition of HO-1 by ZnPP reversed the protective effect of GR on melanocytes against H2O2-induced cytotoxicity and apoptosis. Hydrogen Peroxide 141-145 heme oxygenase 1 Homo sapiens 68-72 31251786-4 2019 In this study we found that HBZ activates expression of Heme Oxygenase 1 (HMOX-1), a component of the oxidative stress response that functions to detoxify free heme. Heme 160-164 heme oxygenase 1 Homo sapiens 56-72 31261663-0 2019 Carbon Monoxide Releasing Molecule-2-Upregulated ROS-Dependent Heme Oxygenase-1 Axis Suppresses Lipopolysaccharide-Induced Airway Inflammation. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 63-79 31251786-4 2019 In this study we found that HBZ activates expression of Heme Oxygenase 1 (HMOX-1), a component of the oxidative stress response that functions to detoxify free heme. Heme 160-164 heme oxygenase 1 Homo sapiens 74-80 31261663-0 2019 Carbon Monoxide Releasing Molecule-2-Upregulated ROS-Dependent Heme Oxygenase-1 Axis Suppresses Lipopolysaccharide-Induced Airway Inflammation. Reactive Oxygen Species 49-52 heme oxygenase 1 Homo sapiens 63-79 31251786-8 2019 Consistent with this model, we found that HMOX-1 is upregulated in HTLV-1-transformed T-cell lines and confers these cells with resistance to heme-induced cytotoxicity. Heme 142-146 heme oxygenase 1 Homo sapiens 42-48 31261663-1 2019 The up-regulation of heme oxygenase-1 (HO-1) is mediated through nicotinamaide adenine dinucleotide phosphate (NADPH) oxidases (Nox) and reactive oxygen species (ROS) generation, which could provide cytoprotection against inflammation. Reactive Oxygen Species 137-160 heme oxygenase 1 Homo sapiens 21-37 31261663-1 2019 The up-regulation of heme oxygenase-1 (HO-1) is mediated through nicotinamaide adenine dinucleotide phosphate (NADPH) oxidases (Nox) and reactive oxygen species (ROS) generation, which could provide cytoprotection against inflammation. Reactive Oxygen Species 137-160 heme oxygenase 1 Homo sapiens 39-43 31208152-8 2019 CHA79 attenuated MG-induced ROS production and enhanced the antioxidant defense including nuclear factor erythroid 2-related factor 2 (Nrf2), HO-1, SOD and GSH. cha79 0-5 heme oxygenase 1 Homo sapiens 142-146 31261663-1 2019 The up-regulation of heme oxygenase-1 (HO-1) is mediated through nicotinamaide adenine dinucleotide phosphate (NADPH) oxidases (Nox) and reactive oxygen species (ROS) generation, which could provide cytoprotection against inflammation. Reactive Oxygen Species 162-165 heme oxygenase 1 Homo sapiens 21-37 31261663-1 2019 The up-regulation of heme oxygenase-1 (HO-1) is mediated through nicotinamaide adenine dinucleotide phosphate (NADPH) oxidases (Nox) and reactive oxygen species (ROS) generation, which could provide cytoprotection against inflammation. Reactive Oxygen Species 162-165 heme oxygenase 1 Homo sapiens 39-43 31261663-2 2019 However, the molecular mechanisms of carbon monoxide-releasing molecule (CORM)-2-induced HO-1 expression in human tracheal smooth muscle cells (HTSMCs) remain unknown. Carbon Monoxide 37-52 heme oxygenase 1 Homo sapiens 89-93 31261663-6 2019 We found that CORM-2 induced HO-1 expression via the activation of protein kinase C (PKC)alpha and proline-rich tyrosine kinase (Pyk2), which was mediated through Nox-derived ROS generation using pharmacological inhibitors or small interfering ribonucleic acids (siRNAs). nicotine 1-N-oxide 163-166 heme oxygenase 1 Homo sapiens 29-33 31261663-6 2019 We found that CORM-2 induced HO-1 expression via the activation of protein kinase C (PKC)alpha and proline-rich tyrosine kinase (Pyk2), which was mediated through Nox-derived ROS generation using pharmacological inhibitors or small interfering ribonucleic acids (siRNAs). Reactive Oxygen Species 175-178 heme oxygenase 1 Homo sapiens 29-33 31261663-10 2019 These results suggested that in HTSMCs, CORM-2 activates PKCalpha/Pyk2-dependent Nox/ROS/ERK1/2/AP-1, leading to HO-1 up-regulation, which suppresses the lipopolysaccharide (LPS)-induced airway inflammation. nicotine 1-N-oxide 81-84 heme oxygenase 1 Homo sapiens 113-117 31261663-10 2019 These results suggested that in HTSMCs, CORM-2 activates PKCalpha/Pyk2-dependent Nox/ROS/ERK1/2/AP-1, leading to HO-1 up-regulation, which suppresses the lipopolysaccharide (LPS)-induced airway inflammation. Reactive Oxygen Species 85-88 heme oxygenase 1 Homo sapiens 113-117 31056260-10 2019 Importantly, CDDP combined with LY294002, inhibitor of phosphoinositide 3-kinase (PI3K)/AKT serine/threonine kinase (AKT) signaling, markedly decreased the expression of NRF2, HO-1, NQO1 and GCLC in drug-resistant cervical cancer cells. Cisplatin 13-17 heme oxygenase 1 Homo sapiens 176-180 31056260-10 2019 Importantly, CDDP combined with LY294002, inhibitor of phosphoinositide 3-kinase (PI3K)/AKT serine/threonine kinase (AKT) signaling, markedly decreased the expression of NRF2, HO-1, NQO1 and GCLC in drug-resistant cervical cancer cells. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 32-40 heme oxygenase 1 Homo sapiens 176-180 31029788-6 2019 Heme oxygenase-1 (HO-1) was induced by quercetin but not its conjugates, but was not implicated in the glucose uptake stimulation since hemin, a classical inducer of HO-1, did not affect glucose metabolism. Quercetin 39-48 heme oxygenase 1 Homo sapiens 0-16 30773786-7 2019 The HO-1 inhibitor tin protoporphyrin IX prevented the anti-senescence action evoked by Ang-(1-7) or recombinant klotho. tin protoporphyrin IX 19-40 heme oxygenase 1 Homo sapiens 4-8 30991050-8 2019 Notably, silencing of Nrf2 (siRNA transfection) significantly diminished CoQ10-mediated anti-melanogenic activity, as evidenced by impaired antioxidant HO-1 gene, uncontrolled ROS generation, and alpha-MSH production following UVA irradiation. coenzyme Q10 73-78 heme oxygenase 1 Homo sapiens 152-156 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Heme 49-53 heme oxygenase 1 Homo sapiens 16-32 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Heme 49-53 heme oxygenase 1 Homo sapiens 34-38 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 16-32 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 34-38 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 16-32 31163581-1 2019 (1) Background: Heme oxygenase-1 (HO-1) degrades heme and generates carbon monoxide (CO), producing various anti-inflammatory, anti-oxidative, and anti-apoptotic effects. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 34-38 31163602-13 2019 extract promotes an oxidative cellular microenvironment resulting in CaCo2 cell death by ferroptosis mediated by HO-1 hyper-expression. caco2 69-74 heme oxygenase 1 Homo sapiens 113-117 30965051-5 2019 Here, we report that Sulforaphane is an inducer of heme oxygenase (HO)-1, a cytoprotective enzyme that catalyzes the degradation of heme through signaling pathways in human monocytes. sulforaphane 21-33 heme oxygenase 1 Homo sapiens 51-72 30841754-4 2019 We now show that increasing total NAD+ content in astrocytes leads to the activation of the transcription factor nuclear factor, erythroid-derived 2, like 2 (Nfe2l2 or Nrf2) and up-regulation of the antioxidant proteins heme oxygenase 1 (HO-1) and sulfiredoxin 1 (SRXN1). NAD 34-38 heme oxygenase 1 Homo sapiens 220-236 31029788-6 2019 Heme oxygenase-1 (HO-1) was induced by quercetin but not its conjugates, but was not implicated in the glucose uptake stimulation since hemin, a classical inducer of HO-1, did not affect glucose metabolism. Quercetin 39-48 heme oxygenase 1 Homo sapiens 18-22 31261522-1 2019 Heme oxygenase-1 (HO-1) is an important catalytic enzyme in heme degradation, which increases during stressful conditions. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 30907046-10 2019 Increased intracellular ROS resulted in nuclear factor kappa B (NF-kappaB) activation and upregulated expression of cyclooxygenase-2 (COX-2), hemeoxygenase-1 (HO-1), interleukin-1beta (IL-1beta), and intercellular adhesion molecule-1 (ICAM-1). Reactive Oxygen Species 24-27 heme oxygenase 1 Homo sapiens 142-163 30884345-0 2019 A novel chalcone derivative, L2H17, ameliorates lipopolysaccharide-induced acute lung injury via upregulating HO-1 activity. Chalcone 8-16 heme oxygenase 1 Homo sapiens 110-114 30884345-0 2019 A novel chalcone derivative, L2H17, ameliorates lipopolysaccharide-induced acute lung injury via upregulating HO-1 activity. l2h17 29-34 heme oxygenase 1 Homo sapiens 110-114 30884345-13 2019 We also found that the inhibitory effect of L2H17 on the inflammatory responses was attenuated by an inhibitor of HO-1 activity, Tin protoporphyrin IX (SnPP). l2h17 44-49 heme oxygenase 1 Homo sapiens 114-118 30884345-13 2019 We also found that the inhibitory effect of L2H17 on the inflammatory responses was attenuated by an inhibitor of HO-1 activity, Tin protoporphyrin IX (SnPP). tin protoporphyrin IX 129-150 heme oxygenase 1 Homo sapiens 114-118 30884345-13 2019 We also found that the inhibitory effect of L2H17 on the inflammatory responses was attenuated by an inhibitor of HO-1 activity, Tin protoporphyrin IX (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 152-156 heme oxygenase 1 Homo sapiens 114-118 30758914-10 2019 ALA treatment decreased ROS production in UPM-exposed fibroblasts via the Nrf2, HO-1, and NQO-1 pathways. Thioctic Acid 0-3 heme oxygenase 1 Homo sapiens 80-84 30758914-10 2019 ALA treatment decreased ROS production in UPM-exposed fibroblasts via the Nrf2, HO-1, and NQO-1 pathways. Reactive Oxygen Species 24-27 heme oxygenase 1 Homo sapiens 80-84 30629288-0 2019 Brusatol inhibits amyloid-beta-induced neurotoxicity in U-251 cells via regulating the Nrf2/HO-1 pathway. brusatol 0-8 heme oxygenase 1 Homo sapiens 92-96 30629288-7 2019 Furthermore, BR induced the Nrf2/HO-1 pathway by inhibiting the PI3K/AKT/mTOR pathway to inhibit neurotoxicity elicited by Abeta. brusatol 13-15 heme oxygenase 1 Homo sapiens 33-37 30817082-7 2019 Hesperidin also obviously upregulates Nrf2 antioxidant pathway and increases the protein expression of HO-1 and NQO1. Hesperidin 0-10 heme oxygenase 1 Homo sapiens 103-107 31261522-1 2019 Heme oxygenase-1 (HO-1) is an important catalytic enzyme in heme degradation, which increases during stressful conditions. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 31137785-0 2019 Heme Oxygenase-1 Inhibition Sensitizes Human Prostate Cancer Cells towards Glucose Deprivation and Metformin-Mediated Cell Death. Glucose 75-82 heme oxygenase 1 Homo sapiens 0-16 31146323-5 2019 We showed that SB could significantly protect SH-SY5Y cells against 6-OHDA-induced cellular damage by inhibiting cell apoptosis and oxidative stress through PI3K/Akt, MAPK, caspase cascade modulation and Nrf2/HO-1 cascade modulation, respectively. stellettin B 15-17 heme oxygenase 1 Homo sapiens 209-213 31137785-4 2019 The present work was directed to determine whether use of a specific inhibitor of HO-1 activity, alone or in combination with metformin, affected metastatic prostate cancer cell viability under different concentrations of glucose. Glucose 222-229 heme oxygenase 1 Homo sapiens 82-86 31137785-7 2019 The activity of HO-1 was inhibited using a selective imidazole-based inhibitor; genes associated with antioxidant systems and cell death were evaluated by qRT-PCR. imidazole 53-62 heme oxygenase 1 Homo sapiens 16-20 31137785-0 2019 Heme Oxygenase-1 Inhibition Sensitizes Human Prostate Cancer Cells towards Glucose Deprivation and Metformin-Mediated Cell Death. Metformin 99-108 heme oxygenase 1 Homo sapiens 0-16 31137785-9 2019 Disrupting the antioxidant HO-1 activity, especially under low glucose concentration, could be an attractive approach to potentiate metformin antineoplastic effects and could provide a biochemical basis for developing HO-1-targeting drugs against solid tumors. Glucose 63-70 heme oxygenase 1 Homo sapiens 27-31 31120979-2 2019 Recently, an increased number of dinucleotide repeats in the HMOX1 gene were reported to associate with development of AKI in cardiac surgery. Dinucleoside Phosphates 33-45 heme oxygenase 1 Homo sapiens 61-66 31137785-9 2019 Disrupting the antioxidant HO-1 activity, especially under low glucose concentration, could be an attractive approach to potentiate metformin antineoplastic effects and could provide a biochemical basis for developing HO-1-targeting drugs against solid tumors. Metformin 132-141 heme oxygenase 1 Homo sapiens 27-31 31193778-4 2019 Resveratrol at concentrations as low as 0.001 muMu increased HO-1 expression as well as membrane cofactor protein (MCP, CD46) and decay-accelerating factor (DAF, CD55) expression with no-effect on CD59. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 61-65 31120979-5 2019 We genotyped 300 patients with severe AKI (KDIGO 2 or 3) and 353 controls without AKI (KDIGO 0) for the guanine-thymine (GTn) repeat in the promoter region of the HMOX1 gene. Guanine 104-111 heme oxygenase 1 Homo sapiens 163-168 31120979-5 2019 We genotyped 300 patients with severe AKI (KDIGO 2 or 3) and 353 controls without AKI (KDIGO 0) for the guanine-thymine (GTn) repeat in the promoter region of the HMOX1 gene. Thymine 112-119 heme oxygenase 1 Homo sapiens 163-168 31120979-5 2019 We genotyped 300 patients with severe AKI (KDIGO 2 or 3) and 353 controls without AKI (KDIGO 0) for the guanine-thymine (GTn) repeat in the promoter region of the HMOX1 gene. Nitroglycerin 121-124 heme oxygenase 1 Homo sapiens 163-168 30944174-6 2019 Here, using hydrogen-deuterium exchange MS, size-exclusion chromatography, and sedimentation velocity, we investigated how these divergent regions impact the dynamics and structure of the apo and heme-bound forms of HO1 and HO2. Heme 196-200 heme oxygenase 1 Homo sapiens 216-219 31049492-0 2019 Wheat alkylresorcinols protect human retinal pigment epithelial cells against H2O2-induced oxidative damage through Akt-dependent Nrf2/HO-1 signaling. wheat alkylresorcinols 0-22 heme oxygenase 1 Homo sapiens 135-139 31049492-0 2019 Wheat alkylresorcinols protect human retinal pigment epithelial cells against H2O2-induced oxidative damage through Akt-dependent Nrf2/HO-1 signaling. Hydrogen Peroxide 78-82 heme oxygenase 1 Homo sapiens 135-139 31049492-3 2019 Moreover, ARs treatment increased NF-E2-related factor 2 (Nrf2) signaling activation, which was evidenced by increased transcription of anti-oxidant responsive genes GCL, NQO1 and HO-1. Alizarin Red S 10-13 heme oxygenase 1 Homo sapiens 180-184 30944174-7 2019 Our results reveal that heme binding to the catalytic cores of HO1 and HO2 causes similar dynamic and structural changes in regions (proximal, distal, and A6 helices) within and linked to the heme pocket. Heme 24-28 heme oxygenase 1 Homo sapiens 63-66 30944174-7 2019 Our results reveal that heme binding to the catalytic cores of HO1 and HO2 causes similar dynamic and structural changes in regions (proximal, distal, and A6 helices) within and linked to the heme pocket. Heme 192-196 heme oxygenase 1 Homo sapiens 63-66 30898432-3 2019 Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme with anti-cytotoxic and anti-inflammatory properties. Heme 136-140 heme oxygenase 1 Homo sapiens 154-158 30084650-5 2019 Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3"-UTR of HO-1, and thereafter decreased HO-1 expression. Homocysteine 0-12 heme oxygenase 1 Homo sapiens 93-97 30084650-5 2019 Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3"-UTR of HO-1, and thereafter decreased HO-1 expression. Homocysteine 0-12 heme oxygenase 1 Homo sapiens 124-128 30084650-5 2019 Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3"-UTR of HO-1, and thereafter decreased HO-1 expression. Homocysteine 14-17 heme oxygenase 1 Homo sapiens 93-97 30084650-5 2019 Homocysteine (Hcy) downregulated HuR expression, reduced the binding of HuR to the 3"-UTR of HO-1, and thereafter decreased HO-1 expression. Homocysteine 14-17 heme oxygenase 1 Homo sapiens 124-128 30084650-6 2019 Administration of the HO-1 inducer cobalt protoporphyrin-IX significantly hindered Hhcy-augmented reactive oxygen species production and renal fibrotic lesions. cobaltiprotoporphyrin 35-59 heme oxygenase 1 Homo sapiens 22-26 30084650-6 2019 Administration of the HO-1 inducer cobalt protoporphyrin-IX significantly hindered Hhcy-augmented reactive oxygen species production and renal fibrotic lesions. Oxygen 107-113 heme oxygenase 1 Homo sapiens 22-26 30576222-0 2019 Klotho protein inhibits H2O2-induced oxidative injury in endothelial cells via regulation of PI3K/AKT/Nrf2/HO-1 pathways. Hydrogen Peroxide 24-28 heme oxygenase 1 Homo sapiens 107-111 30576222-7 2019 Klotho protein also activated phosphorylation of protein kinase B (AKT), whereas the addition of LY294002, a pharmacological inhibitor of phosphatidylinositol 3-kinase (PI3K), blocked Klotho-protein-induced Nrf2/HO-1 activation and cytoprotection. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 97-105 heme oxygenase 1 Homo sapiens 212-216 30576222-8 2019 Klotho protein enhanced the antioxidant defense ability of the cells by activating the PI3K/AKT pathway, which upregulated the expression of Nrf2/HO-1, thereby inhibiting H2O2-induced oxidative damage. Hydrogen Peroxide 171-175 heme oxygenase 1 Homo sapiens 146-150 30653942-0 2019 Enhancing responsiveness of pancreatic cancer cells to gemcitabine treatment under hypoxia by heme oxygenase-1 inhibition. gemcitabine 55-66 heme oxygenase 1 Homo sapiens 94-110 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. zinc protoporphyrin 128-147 heme oxygenase 1 Homo sapiens 86-90 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. zinc protoporphyrin 128-147 heme oxygenase 1 Homo sapiens 86-90 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. zinc protoporphyrin 128-147 heme oxygenase 1 Homo sapiens 86-90 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. S-Nitroso-N-propionyl-D,L-penicillamine 172-176 heme oxygenase 1 Homo sapiens 86-90 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. S-Nitroso-N-propionyl-D,L-penicillamine 172-176 heme oxygenase 1 Homo sapiens 86-90 30653942-5 2019 Our results showed that hypoxia upregulates the expression of HO-1 in PDAC cells, and HO-1 inhibition using the HO-1 inhibitors zinc protoporphyrin, tin protoporphyrin IX (SnPP), and HO-1 knockout using CRISPR/Cas9 suppresses the proliferation of PDAC cells under hypoxia and sensitize them to gemcitabine under in vitro conditions. S-Nitroso-N-propionyl-D,L-penicillamine 172-176 heme oxygenase 1 Homo sapiens 86-90 30653942-7 2019 Mechanistically, inhibition of HO-1 increased the production of reactive oxygen species as demonstrated by increased dihydroethidium, and Mitosox, disrupted glutathione cycle, and enhanced apoptosis. Reactive Oxygen Species 64-87 heme oxygenase 1 Homo sapiens 31-35 30653942-7 2019 Mechanistically, inhibition of HO-1 increased the production of reactive oxygen species as demonstrated by increased dihydroethidium, and Mitosox, disrupted glutathione cycle, and enhanced apoptosis. dihydroethidium 117-132 heme oxygenase 1 Homo sapiens 31-35 30653942-7 2019 Mechanistically, inhibition of HO-1 increased the production of reactive oxygen species as demonstrated by increased dihydroethidium, and Mitosox, disrupted glutathione cycle, and enhanced apoptosis. 5-(6'-triphenylphosphoniumhexyl)-5,6-dihydro-6-phenyl-3,8-phenanthridinediammine 138-145 heme oxygenase 1 Homo sapiens 31-35 30653942-7 2019 Mechanistically, inhibition of HO-1 increased the production of reactive oxygen species as demonstrated by increased dihydroethidium, and Mitosox, disrupted glutathione cycle, and enhanced apoptosis. Glutathione 157-168 heme oxygenase 1 Homo sapiens 31-35 30653942-9 2019 In addition, inhibiting HO-1 reduced expression of stemness markers (CD133, and CD44) as compared to control or gemcitabine. gemcitabine 112-123 heme oxygenase 1 Homo sapiens 24-28 30807827-10 2019 iHAEs showed relatively high resistance to ROS stimulation, which can be attributed to the high SOD2 expression and up-regulation of Nrf2, HO-1 after x-rays exposure. Reactive Oxygen Species 43-46 heme oxygenase 1 Homo sapiens 139-143 30924710-7 2019 Interestingly, antioxidant enzymes, including heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase, were prominently induced by glyceollins in p53 wild-type xenografts, compared with p53 null xenografts. glyceollin 135-146 heme oxygenase 1 Homo sapiens 46-62 30924710-8 2019 These results suggest that a high dose of glyceollins possibly promotes the growth of p53 wild-type colon cancer through activation of the Nrf2-mediated signaling pathway and, in particular, strong induction of HO-1 expression. glyceollin 42-53 heme oxygenase 1 Homo sapiens 211-215 31052354-7 2019 The ectopic expression of HO-1 recovered YD-15 cells from TW-37-induced apoptosis by reducing intracellular levels of ROS. Reactive Oxygen Species 118-121 heme oxygenase 1 Homo sapiens 26-30 31010200-0 2019 Marine Compound 3-bromo-4,5-dihydroxybenzaldehyde Protects Skin Cells against Oxidative Damage via the Nrf2/HO-1 Pathway. 3-bromo-4,5-dihydroxybenzaldehyde 16-49 heme oxygenase 1 Homo sapiens 108-112 31010200-1 2019 In this study, we aimed to illustrate the potential bio-effects of 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on the antioxidant/cytoprotective enzyme heme oxygenase-1 (HO-1) in keratinocytes. 3-bromo-4,5-dihydroxybenzaldehyde 67-100 heme oxygenase 1 Homo sapiens 150-166 31010200-5 2019 Moreover, 3-BDB induced the phosphorylation of ERK and Akt, while inhibitors of ERK and Akt abrogated the 3-BDB-enhanced levels of HO-1 and Nrf2. 3-bromo-4,5-dihydroxybenzaldehyde 106-111 heme oxygenase 1 Homo sapiens 131-135 31010200-1 2019 In this study, we aimed to illustrate the potential bio-effects of 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on the antioxidant/cytoprotective enzyme heme oxygenase-1 (HO-1) in keratinocytes. 3-bromo-4,5-dihydroxybenzaldehyde 67-100 heme oxygenase 1 Homo sapiens 168-172 31010200-8 2019 The present results indicate that 3-BDB activated Nrf2 signaling cascades in keratinocytes, which was mediated by ERK and Akt, upregulated HO-1, and induced cytoprotective effects against oxidative stress. 3-bromo-4,5-dihydroxybenzaldehyde 34-39 heme oxygenase 1 Homo sapiens 139-143 31010200-1 2019 In this study, we aimed to illustrate the potential bio-effects of 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on the antioxidant/cytoprotective enzyme heme oxygenase-1 (HO-1) in keratinocytes. 3-bromo-4,5-dihydroxybenzaldehyde 102-107 heme oxygenase 1 Homo sapiens 150-166 31010200-1 2019 In this study, we aimed to illustrate the potential bio-effects of 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on the antioxidant/cytoprotective enzyme heme oxygenase-1 (HO-1) in keratinocytes. 3-bromo-4,5-dihydroxybenzaldehyde 102-107 heme oxygenase 1 Homo sapiens 168-172 31010200-2 2019 The antioxidant effects of 3-BDB were examined via reverse transcription PCR, Western blotting, HO-1 activity assay, and immunocytochemistry. 3-bromo-4,5-dihydroxybenzaldehyde 27-32 heme oxygenase 1 Homo sapiens 96-100 30876940-6 2019 Hemin or ZnPP was combined to regulate heme oxygenase-1 (HO-1), and a pathway inhibitor was added to measure changes in the JNK/AP-1 signaling pathway. Hemin 0-5 heme oxygenase 1 Homo sapiens 39-55 30851935-3 2019 The cell oxidative stress models were incubated with Hemin (an inducer of HO-1), then, the HTR8/SVneo cells were transfected by ZO-1 small interfering RNA (siRNA). Hemin 53-58 heme oxygenase 1 Homo sapiens 74-78 30851935-6 2019 In both the trophoblastic and HUVEC oxidative stress models, HO-1 ZO-1 and occludin were increased incubated with Hemin. Hemin 114-119 heme oxygenase 1 Homo sapiens 61-65 30995787-1 2019 Reactive oxygen species (ROS) induce nuclear factor erythroid 2-related factor 2 (Nrf2) activation as an adaptive defense mechanism, determining the synthesis of antioxidant molecules, including heme-oxygenase-1 (HO-1). Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 195-211 30995787-1 2019 Reactive oxygen species (ROS) induce nuclear factor erythroid 2-related factor 2 (Nrf2) activation as an adaptive defense mechanism, determining the synthesis of antioxidant molecules, including heme-oxygenase-1 (HO-1). Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 213-217 30995787-1 2019 Reactive oxygen species (ROS) induce nuclear factor erythroid 2-related factor 2 (Nrf2) activation as an adaptive defense mechanism, determining the synthesis of antioxidant molecules, including heme-oxygenase-1 (HO-1). Reactive Oxygen Species 25-28 heme oxygenase 1 Homo sapiens 195-211 30995787-1 2019 Reactive oxygen species (ROS) induce nuclear factor erythroid 2-related factor 2 (Nrf2) activation as an adaptive defense mechanism, determining the synthesis of antioxidant molecules, including heme-oxygenase-1 (HO-1). Reactive Oxygen Species 25-28 heme oxygenase 1 Homo sapiens 213-217 30995787-2 2019 HO-1 protects cells against oxidative injury, degrading free heme and inhibiting ROS production. Heme 61-65 heme oxygenase 1 Homo sapiens 0-4 30995787-2 2019 HO-1 protects cells against oxidative injury, degrading free heme and inhibiting ROS production. Reactive Oxygen Species 81-84 heme oxygenase 1 Homo sapiens 0-4 31013932-0 2019 Protective Effect of Phloroglucinol on Oxidative Stress-Induced DNA Damage and Apoptosis through Activation of the Nrf2/HO-1 Signaling Pathway in HaCaT Human Keratinocytes. Phloroglucinol 21-35 heme oxygenase 1 Homo sapiens 120-124 30876940-6 2019 Hemin or ZnPP was combined to regulate heme oxygenase-1 (HO-1), and a pathway inhibitor was added to measure changes in the JNK/AP-1 signaling pathway. Hemin 0-5 heme oxygenase 1 Homo sapiens 57-61 31013932-4 2019 The results showed that PG significantly inhibited the H2O2-induced growth inhibition in HaCaT cells, which was associated with increased expression of heme oxygenase-1 (HO-1) by the activation of nuclear factor erythroid 2-related factor-2 (Nrf2). Phloroglucinol 24-26 heme oxygenase 1 Homo sapiens 152-168 31013932-4 2019 The results showed that PG significantly inhibited the H2O2-induced growth inhibition in HaCaT cells, which was associated with increased expression of heme oxygenase-1 (HO-1) by the activation of nuclear factor erythroid 2-related factor-2 (Nrf2). Phloroglucinol 24-26 heme oxygenase 1 Homo sapiens 170-174 30784878-3 2019 Under these conditions, HO-1 exerts its strong cytoprotective activities and plays a crucial role in stimulating cell survival by removing the pro-oxidant heme and by producing carbon monoxide and biliverdin (promptly reduced to bilirubin). Heme 155-159 heme oxygenase 1 Homo sapiens 24-28 31013932-4 2019 The results showed that PG significantly inhibited the H2O2-induced growth inhibition in HaCaT cells, which was associated with increased expression of heme oxygenase-1 (HO-1) by the activation of nuclear factor erythroid 2-related factor-2 (Nrf2). Hydrogen Peroxide 55-59 heme oxygenase 1 Homo sapiens 152-168 31013932-4 2019 The results showed that PG significantly inhibited the H2O2-induced growth inhibition in HaCaT cells, which was associated with increased expression of heme oxygenase-1 (HO-1) by the activation of nuclear factor erythroid 2-related factor-2 (Nrf2). Hydrogen Peroxide 55-59 heme oxygenase 1 Homo sapiens 170-174 31013932-8 2019 However, the inhibition of HO-1 function using zinc protoporphyrin, a HO-1 inhibitor, markedly attenuated these protective effects of PG against H2O2. zinc protoporphyrin 47-66 heme oxygenase 1 Homo sapiens 27-31 31013932-8 2019 However, the inhibition of HO-1 function using zinc protoporphyrin, a HO-1 inhibitor, markedly attenuated these protective effects of PG against H2O2. zinc protoporphyrin 47-66 heme oxygenase 1 Homo sapiens 70-74 31013932-8 2019 However, the inhibition of HO-1 function using zinc protoporphyrin, a HO-1 inhibitor, markedly attenuated these protective effects of PG against H2O2. Phloroglucinol 134-136 heme oxygenase 1 Homo sapiens 27-31 31013932-8 2019 However, the inhibition of HO-1 function using zinc protoporphyrin, a HO-1 inhibitor, markedly attenuated these protective effects of PG against H2O2. Phloroglucinol 134-136 heme oxygenase 1 Homo sapiens 70-74 31013932-8 2019 However, the inhibition of HO-1 function using zinc protoporphyrin, a HO-1 inhibitor, markedly attenuated these protective effects of PG against H2O2. Hydrogen Peroxide 145-149 heme oxygenase 1 Homo sapiens 27-31 31013932-9 2019 Overall, our results suggest that PG is able to protect HaCaT keratinocytes against oxidative stress-induced DNA damage and apoptosis through activating the Nrf2/HO-1 signaling pathway. Phloroglucinol 34-36 heme oxygenase 1 Homo sapiens 162-166 30965670-3 2019 Gene silencing (siRNA-mediated) was performed to examine the responses of Nrf2-target genes (i.e., heme oxygenase-1, NAD(P)H:quinone oxidoreductase1) to siRNA depletion of Nrf2 in MPP+-induced dopaminergic cell death. mangion-purified polysaccharide (Candida albicans) 180-184 heme oxygenase 1 Homo sapiens 99-148 31089407-4 2019 It was found that HEMA stimulation promoted nuclear translocation of Nrf2 and increased Nrf2 and heme oxygenase-1 (HO-1) expression, which was further enhanced by Nrf2 activator tert-butylhydroquinone (tBHQ), but suppressed by Nrf2 inhibitor ML385. hydroxyethyl methacrylate 18-22 heme oxygenase 1 Homo sapiens 97-113 31089407-4 2019 It was found that HEMA stimulation promoted nuclear translocation of Nrf2 and increased Nrf2 and heme oxygenase-1 (HO-1) expression, which was further enhanced by Nrf2 activator tert-butylhydroquinone (tBHQ), but suppressed by Nrf2 inhibitor ML385. 2-tert-butylhydroquinone 202-206 heme oxygenase 1 Homo sapiens 97-113 31089407-4 2019 It was found that HEMA stimulation promoted nuclear translocation of Nrf2 and increased Nrf2 and heme oxygenase-1 (HO-1) expression, which was further enhanced by Nrf2 activator tert-butylhydroquinone (tBHQ), but suppressed by Nrf2 inhibitor ML385. ML385 242-247 heme oxygenase 1 Homo sapiens 97-113 30974805-0 2019 Ethanol-Mediated Stress Promotes Autophagic Survival and Aggressiveness of Colon Cancer Cells via Activation of Nrf2/HO-1 Pathway. Ethanol 0-7 heme oxygenase 1 Homo sapiens 117-121 30974805-6 2019 EtOH treatment also upregulates the antioxidant enzymes SOD, catalase, and heme oxygenase (HO-1) and promotes the nuclear translocation of both Nrf2 and HO-1. Ethanol 0-4 heme oxygenase 1 Homo sapiens 91-95 30974805-6 2019 EtOH treatment also upregulates the antioxidant enzymes SOD, catalase, and heme oxygenase (HO-1) and promotes the nuclear translocation of both Nrf2 and HO-1. Ethanol 0-4 heme oxygenase 1 Homo sapiens 153-157 30974805-8 2019 Nrf2 silencing or preventing HO-1 nuclear translocation by the protease inhibitor E64d abrogates the EtOH-induced increase in the antioxidant enzyme levels as well as the migration markers. aloxistatin 82-86 heme oxygenase 1 Homo sapiens 29-33 30974805-8 2019 Nrf2 silencing or preventing HO-1 nuclear translocation by the protease inhibitor E64d abrogates the EtOH-induced increase in the antioxidant enzyme levels as well as the migration markers. Ethanol 101-105 heme oxygenase 1 Homo sapiens 29-33 30974805-9 2019 Taken together, our results suggest that EtOH mediates both the activation of Nrf2 and HO-1 to sustain colon cancer cell survival, thus leading to the acquisition of a more aggressive phenotype. Ethanol 41-45 heme oxygenase 1 Homo sapiens 87-91 30594636-8 2019 This led to HO-1 protein downregulation in H2O2-treated human pancreatic beta-cells, which was also observed in NRF2-silenced beta-cells. Hydrogen Peroxide 43-47 heme oxygenase 1 Homo sapiens 12-16 30784878-3 2019 Under these conditions, HO-1 exerts its strong cytoprotective activities and plays a crucial role in stimulating cell survival by removing the pro-oxidant heme and by producing carbon monoxide and biliverdin (promptly reduced to bilirubin). Carbon Monoxide 177-192 heme oxygenase 1 Homo sapiens 24-28 30784878-3 2019 Under these conditions, HO-1 exerts its strong cytoprotective activities and plays a crucial role in stimulating cell survival by removing the pro-oxidant heme and by producing carbon monoxide and biliverdin (promptly reduced to bilirubin). Biliverdine 197-207 heme oxygenase 1 Homo sapiens 24-28 30784878-3 2019 Under these conditions, HO-1 exerts its strong cytoprotective activities and plays a crucial role in stimulating cell survival by removing the pro-oxidant heme and by producing carbon monoxide and biliverdin (promptly reduced to bilirubin). Bilirubin 229-238 heme oxygenase 1 Homo sapiens 24-28 30784878-5 2019 Development of non-competitive HO-1 inhibitors dates back in 2002 with the discovery of Azalanstat. azalanstat 88-98 heme oxygenase 1 Homo sapiens 31-35 30883732-0 2019 Crystal structure of a NADPH-cytochrome P450 oxidoreductase (CYPOR) and heme oxygenase 1 fusion protein implies a conformational change in CYPOR upon NADPH/NADP+ binding. NADP 156-161 heme oxygenase 1 Homo sapiens 72-88 30769048-0 2019 Mangiferin alleviates arsenic induced oxidative lung injury via upregulation of the Nrf2-HO1 axis. mangiferin 0-10 heme oxygenase 1 Homo sapiens 89-92 30769048-0 2019 Mangiferin alleviates arsenic induced oxidative lung injury via upregulation of the Nrf2-HO1 axis. Arsenic 22-29 heme oxygenase 1 Homo sapiens 89-92 30883732-1 2019 Heme oxygenase-1 (HMOX1) catalyzes heme degradation utilizing reducing equivalents supplied from NADPH-cytochrome P450 reductase (CYPOR). Heme 35-39 heme oxygenase 1 Homo sapiens 0-16 30883732-1 2019 Heme oxygenase-1 (HMOX1) catalyzes heme degradation utilizing reducing equivalents supplied from NADPH-cytochrome P450 reductase (CYPOR). Heme 35-39 heme oxygenase 1 Homo sapiens 18-23 30883732-2 2019 Recently, we determined the complex structure of NADP+ -bound open-conformation stabilized CYPOR and heme-HMOX1, but the resolution was limited to 4.3 A. NADP 49-54 heme oxygenase 1 Homo sapiens 106-111 30883732-6 2019 As a result of this change, the FMN-binding domain of CYPOR approaches heme-bound HMOX1 upon NADP+ binding to enhance the electron-transfer efficiency from FMN to heme. Heme 71-75 heme oxygenase 1 Homo sapiens 82-87 30883732-6 2019 As a result of this change, the FMN-binding domain of CYPOR approaches heme-bound HMOX1 upon NADP+ binding to enhance the electron-transfer efficiency from FMN to heme. NADP 93-98 heme oxygenase 1 Homo sapiens 82-87 30883732-6 2019 As a result of this change, the FMN-binding domain of CYPOR approaches heme-bound HMOX1 upon NADP+ binding to enhance the electron-transfer efficiency from FMN to heme. Flavin Mononucleotide 32-35 heme oxygenase 1 Homo sapiens 82-87 30883732-6 2019 As a result of this change, the FMN-binding domain of CYPOR approaches heme-bound HMOX1 upon NADP+ binding to enhance the electron-transfer efficiency from FMN to heme. Heme 163-167 heme oxygenase 1 Homo sapiens 82-87 30256411-0 2019 Heme oxygenase-1 reduces the sensitivity to imatinib through nonselective activation of histone deacetylases in chronic myeloid leukemia. Imatinib Mesylate 44-52 heme oxygenase 1 Homo sapiens 0-16 30594624-0 2019 Nitric oxide protected against NADPH oxidase-derived superoxide generation in vascular endothelium: Critical role for heme oxygenase-1. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 118-134 30594624-2 2019 Nitric oxide (NO), previously viewed as a key regulator of cardiovascular function, is identified to induce expression of heme oxygenase-1 (HO-1), an antioxidant enzyme in blood vessels. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 122-138 30594624-2 2019 Nitric oxide (NO), previously viewed as a key regulator of cardiovascular function, is identified to induce expression of heme oxygenase-1 (HO-1), an antioxidant enzyme in blood vessels. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 140-144 30594624-4 2019 In vascular endothelial cells, NO donor (diethylenetriamine/nitric oxide adduct) significantly attenuated NADPH oxidase-derived O2- generation via a HO-1-dependent mechanism. diethylenetriamine 41-59 heme oxygenase 1 Homo sapiens 149-153 30594624-4 2019 In vascular endothelial cells, NO donor (diethylenetriamine/nitric oxide adduct) significantly attenuated NADPH oxidase-derived O2- generation via a HO-1-dependent mechanism. Nitric Oxide 60-72 heme oxygenase 1 Homo sapiens 149-153 30594624-4 2019 In vascular endothelial cells, NO donor (diethylenetriamine/nitric oxide adduct) significantly attenuated NADPH oxidase-derived O2- generation via a HO-1-dependent mechanism. Superoxides 128-130 heme oxygenase 1 Homo sapiens 149-153 30594624-7 2019 In addition, NO inhibited H2O2-induced endothelial dysfunction through activation of HO-1. Hydrogen Peroxide 26-30 heme oxygenase 1 Homo sapiens 85-89 30256411-8 2019 These findings related to the inhibitory effects of high HO-1 expression on the reactive oxygen species (ROS) signaling pathway that negatively regulated HDACs. Reactive Oxygen Species 80-103 heme oxygenase 1 Homo sapiens 57-61 30256411-8 2019 These findings related to the inhibitory effects of high HO-1 expression on the reactive oxygen species (ROS) signaling pathway that negatively regulated HDACs. Reactive Oxygen Species 105-108 heme oxygenase 1 Homo sapiens 57-61 30256411-9 2019 Increased expression of HO-1 activated HDACs by inhibiting ROS production. Reactive Oxygen Species 59-62 heme oxygenase 1 Homo sapiens 24-28 31056142-8 2019 Sulforaphane induced activation and nuclear translocation of NRF2 in HUVECs, inducing heme oxygenase 1. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 86-102 30652259-0 2019 Carvacrol Depends on Heme Oxygenase-1 (HO-1) to Exert Antioxidant, Anti-inflammatory, and Mitochondria-Related Protection in the Human Neuroblastoma SH-SY5Y Cells Line Exposed to Hydrogen Peroxide. carvacrol 0-9 heme oxygenase 1 Homo sapiens 21-37 30652259-0 2019 Carvacrol Depends on Heme Oxygenase-1 (HO-1) to Exert Antioxidant, Anti-inflammatory, and Mitochondria-Related Protection in the Human Neuroblastoma SH-SY5Y Cells Line Exposed to Hydrogen Peroxide. carvacrol 0-9 heme oxygenase 1 Homo sapiens 39-43 30652259-0 2019 Carvacrol Depends on Heme Oxygenase-1 (HO-1) to Exert Antioxidant, Anti-inflammatory, and Mitochondria-Related Protection in the Human Neuroblastoma SH-SY5Y Cells Line Exposed to Hydrogen Peroxide. Hydrogen Peroxide 179-196 heme oxygenase 1 Homo sapiens 39-43 30652259-10 2019 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 microM) suppressed the preventive effects caused by CAR regarding mitochondrial function and inflammation. zinc protoporphyrin 56-78 heme oxygenase 1 Homo sapiens 22-38 30652259-10 2019 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 microM) suppressed the preventive effects caused by CAR regarding mitochondrial function and inflammation. zinc protoporphyrin 56-78 heme oxygenase 1 Homo sapiens 40-44 30652259-10 2019 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 microM) suppressed the preventive effects caused by CAR regarding mitochondrial function and inflammation. zinc protoporphyrin 80-87 heme oxygenase 1 Homo sapiens 22-38 30652259-10 2019 The inhibition of the heme oxygenase-1 (HO-1) enzyme by zinc protoporphyrin IX (ZnPP IX, 10 microM) suppressed the preventive effects caused by CAR regarding mitochondrial function and inflammation. zinc protoporphyrin 80-87 heme oxygenase 1 Homo sapiens 40-44 30652259-11 2019 Thus, it is suggested that CAR caused cytoprotective effects by an HO-1-dependent manner in SH-SY5Y cells exposed to H2O2. carvacrol 27-30 heme oxygenase 1 Homo sapiens 67-71 30652259-11 2019 Thus, it is suggested that CAR caused cytoprotective effects by an HO-1-dependent manner in SH-SY5Y cells exposed to H2O2. Hydrogen Peroxide 117-121 heme oxygenase 1 Homo sapiens 67-71 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Carbon Monoxide 51-66 heme oxygenase 1 Homo sapiens 20-36 30826469-4 2019 Moreover, Nrf2 translocation appeared both in GD 12.5 d and GD 18.5 d. In conclusion, DON-induced ROS accumulation may cause maternal liver damage in the initial stages, but the related stimulation of Nrf2/HO-1 pathway improves the removal of ROS and decreases the level of oxidative stress thereby protecting the liver damage. ros 243-246 heme oxygenase 1 Homo sapiens 206-210 30826469-3 2019 DON slightly increased the levels of ALT and AST in GD12.5 d instead of GD18.5 d. Oxidative stress and anti-oxidization system were both found to be activated in the experiment which marked by ROS, MDA and GSH increasing, especially on GD12.5 d. The levels of HO-1 were significantly increased by DON exposure at different two time points. deoxynivalenol 0-3 heme oxygenase 1 Homo sapiens 260-264 30826469-4 2019 Moreover, Nrf2 translocation appeared both in GD 12.5 d and GD 18.5 d. In conclusion, DON-induced ROS accumulation may cause maternal liver damage in the initial stages, but the related stimulation of Nrf2/HO-1 pathway improves the removal of ROS and decreases the level of oxidative stress thereby protecting the liver damage. deoxynivalenol 86-89 heme oxygenase 1 Homo sapiens 206-210 30826469-4 2019 Moreover, Nrf2 translocation appeared both in GD 12.5 d and GD 18.5 d. In conclusion, DON-induced ROS accumulation may cause maternal liver damage in the initial stages, but the related stimulation of Nrf2/HO-1 pathway improves the removal of ROS and decreases the level of oxidative stress thereby protecting the liver damage. ros 98-101 heme oxygenase 1 Homo sapiens 206-210 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Carbon Monoxide 51-66 heme oxygenase 1 Homo sapiens 38-42 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. NADP 137-180 heme oxygenase 1 Homo sapiens 20-36 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. NADP 137-180 heme oxygenase 1 Homo sapiens 38-42 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. NADP 182-187 heme oxygenase 1 Homo sapiens 20-36 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. NADP 182-187 heme oxygenase 1 Homo sapiens 38-42 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. nicotine 1-N-oxide 199-202 heme oxygenase 1 Homo sapiens 20-36 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. nicotine 1-N-oxide 199-202 heme oxygenase 1 Homo sapiens 38-42 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Reactive Oxygen Species 208-231 heme oxygenase 1 Homo sapiens 20-36 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Reactive Oxygen Species 208-231 heme oxygenase 1 Homo sapiens 38-42 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Reactive Oxygen Species 233-236 heme oxygenase 1 Homo sapiens 20-36 30934992-1 2019 The upregulation of heme oxygenase-1 (HO-1) by the carbon monoxide-releasing molecule (CORM)-2 may be mediated through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidases [Nox] and reactive oxygen species (ROS) generation, which could provide cytoprotection against various cellular injuries. Reactive Oxygen Species 233-236 heme oxygenase 1 Homo sapiens 38-42 31237164-10 2019 HMOX1 and SOD1 were downregulated after both 24-hour PAH treatments. Polycyclic Aromatic Hydrocarbons 53-56 heme oxygenase 1 Homo sapiens 0-5 30769116-9 2019 Genetic inhibition of Nrf2 greatly decreased the protective effect of edaravone against cell apoptosis and cytotoxicity induced by Abeta25-35, accompanied by decreases in SOD and HO-1 expression. Edaravone 70-79 heme oxygenase 1 Homo sapiens 179-183 30196486-10 2019 Treatment with 10 muM BA increased the mRNA levels of all three genes at 1-4 h in DU-145 cells and HMOX1 and GCLC in MEF WT cells. boric acid 22-24 heme oxygenase 1 Homo sapiens 99-104 30783417-6 2019 Western blot assay showed that allicin could suppress the expression of nuclear factor erythroid 2-related factor 2 (NRF2) and heme oxygenase 1. allicin 31-38 heme oxygenase 1 Homo sapiens 127-143 30824385-8 2019 Sulfasalazine is known to inhibit nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) and upregulate heme-oxygenase 1 (HO-1) thus we explored the effect of these transcription factors on sFlt-1 secretion from human cytotrophoblasts. Sulfasalazine 0-13 heme oxygenase 1 Homo sapiens 119-135 30824385-8 2019 Sulfasalazine is known to inhibit nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) and upregulate heme-oxygenase 1 (HO-1) thus we explored the effect of these transcription factors on sFlt-1 secretion from human cytotrophoblasts. Sulfasalazine 0-13 heme oxygenase 1 Homo sapiens 137-141 30881359-0 2019 Plant-Derived Polyphenols Modulate Human Dendritic Cell Metabolism and Immune Function via AMPK-Dependent Induction of Heme Oxygenase-1. Polyphenols 14-25 heme oxygenase 1 Homo sapiens 119-135 30881359-7 2019 Previously we have reported that both carnosol and curcumin can regulate the maturation and function of human DC through upregulation of the immunomodulatory enzyme, Heme Oxygenase-1 (HO-1). carnosol 38-46 heme oxygenase 1 Homo sapiens 166-182 30881359-7 2019 Previously we have reported that both carnosol and curcumin can regulate the maturation and function of human DC through upregulation of the immunomodulatory enzyme, Heme Oxygenase-1 (HO-1). carnosol 38-46 heme oxygenase 1 Homo sapiens 184-188 30881359-7 2019 Previously we have reported that both carnosol and curcumin can regulate the maturation and function of human DC through upregulation of the immunomodulatory enzyme, Heme Oxygenase-1 (HO-1). Curcumin 51-59 heme oxygenase 1 Homo sapiens 166-182 30881359-7 2019 Previously we have reported that both carnosol and curcumin can regulate the maturation and function of human DC through upregulation of the immunomodulatory enzyme, Heme Oxygenase-1 (HO-1). Curcumin 51-59 heme oxygenase 1 Homo sapiens 184-188 30881359-8 2019 Here we also demonstrate that the induction of HO-1 by polyphenols in human DC is dependent on their activation of AMPK. Polyphenols 55-66 heme oxygenase 1 Homo sapiens 47-51 30881359-10 2019 This study therefore describes a novel relationship between metabolic signaling via AMPK and HO-1 induction by carnosol and curcumin in human DC, and characterizes the effects of these polyphenols on DC immunometabolism for the first time. carnosol 111-119 heme oxygenase 1 Homo sapiens 93-97 30881359-10 2019 This study therefore describes a novel relationship between metabolic signaling via AMPK and HO-1 induction by carnosol and curcumin in human DC, and characterizes the effects of these polyphenols on DC immunometabolism for the first time. Curcumin 124-132 heme oxygenase 1 Homo sapiens 93-97 30269355-8 2019 Moreover, TQ enhanced the activation of Nrf2/heme oxygenase 1 (HO-1) signaling pathway in H2 O2 -induced ARPE cells. thymoquinone 10-12 heme oxygenase 1 Homo sapiens 40-61 30269355-8 2019 Moreover, TQ enhanced the activation of Nrf2/heme oxygenase 1 (HO-1) signaling pathway in H2 O2 -induced ARPE cells. thymoquinone 10-12 heme oxygenase 1 Homo sapiens 63-67 30269355-8 2019 Moreover, TQ enhanced the activation of Nrf2/heme oxygenase 1 (HO-1) signaling pathway in H2 O2 -induced ARPE cells. Hydrogen Peroxide 90-95 heme oxygenase 1 Homo sapiens 40-61 30270549-8 2019 Furthermore, higenamine elevated the expression levels of p-Akt, heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2). higenamine 13-23 heme oxygenase 1 Homo sapiens 65-81 30269355-8 2019 Moreover, TQ enhanced the activation of Nrf2/heme oxygenase 1 (HO-1) signaling pathway in H2 O2 -induced ARPE cells. Hydrogen Peroxide 90-95 heme oxygenase 1 Homo sapiens 63-67 30270549-8 2019 Furthermore, higenamine elevated the expression levels of p-Akt, heme oxygenase-1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2). higenamine 13-23 heme oxygenase 1 Homo sapiens 83-87 30270549-10 2019 These findings indicated that higenamine protects neuronal cells against OGD/R-induced injury by regulating the Akt and Nrf2/HO-1-signaling pathways. higenamine 30-40 heme oxygenase 1 Homo sapiens 125-129 30269355-10 2019 These results suggested that TQ protected ARPE cells from H2 O2 -induced oxidative stress and apoptosis via the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 58-63 heme oxygenase 1 Homo sapiens 117-121 30604480-6 2019 Furthermore, activation of nuclear factor kappaB (NF-kappaB), which was induced by IL-1beta, was also inhibited by SA through the pathway of nuclear factor-erythroid 2-related factor-2 (Nrf2)/heme oxygenase 1. sinapinic acid 115-117 heme oxygenase 1 Homo sapiens 192-208 30419276-0 2019 Nepeta angustifolia attenuates responses to vascular inflammation in high glucose-induced human umbilical vein endothelial cells through heme oxygenase-1 induction. Glucose 74-81 heme oxygenase 1 Homo sapiens 137-153 30709701-0 2019 Oxyresveratrol protects human lens epithelial cells against hydrogen peroxide-induced oxidative stress and apoptosis by activation of Akt/HO-1 pathway. puag-haad 0-14 heme oxygenase 1 Homo sapiens 138-142 30747216-9 2019 Furthermore, an increase in H2S production enhanced the nuclear accumulation of Nrf2 and expression of its downstream targets, heme oxygenase-1 and superoxide dismutase, as well as HIF-1alpha. Hydrogen Sulfide 28-31 heme oxygenase 1 Homo sapiens 127-168 30709701-0 2019 Oxyresveratrol protects human lens epithelial cells against hydrogen peroxide-induced oxidative stress and apoptosis by activation of Akt/HO-1 pathway. Hydrogen Peroxide 60-77 heme oxygenase 1 Homo sapiens 138-142 30709701-9 2019 Oxy increased p-Akt and HO-1 expressions in H2O2-stimulated HLECs. Hydrogen Peroxide 44-48 heme oxygenase 1 Homo sapiens 24-28 30709701-10 2019 Akt and HO-1 expressions form a regulatory axis and Oxy activated the Akt/HO-1 pathway in H2O2-stimulated HLECs. Hydrogen Peroxide 90-94 heme oxygenase 1 Homo sapiens 8-12 30709701-10 2019 Akt and HO-1 expressions form a regulatory axis and Oxy activated the Akt/HO-1 pathway in H2O2-stimulated HLECs. Hydrogen Peroxide 90-94 heme oxygenase 1 Homo sapiens 74-78 30709701-11 2019 Inhibition of the Akt/HO-1 pathway by LY294002 or ZnPP attenuated the effects of Oxy on oxidative stress and apoptosis in H2O2-stimulated HLECs. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 38-46 heme oxygenase 1 Homo sapiens 22-26 30709701-11 2019 Inhibition of the Akt/HO-1 pathway by LY294002 or ZnPP attenuated the effects of Oxy on oxidative stress and apoptosis in H2O2-stimulated HLECs. zinc protoporphyrin 50-54 heme oxygenase 1 Homo sapiens 22-26 30709701-11 2019 Inhibition of the Akt/HO-1 pathway by LY294002 or ZnPP attenuated the effects of Oxy on oxidative stress and apoptosis in H2O2-stimulated HLECs. Hydrogen Peroxide 122-126 heme oxygenase 1 Homo sapiens 22-26 30709701-12 2019 In conclusion, Oxy protected H2O2-induced oxidative stress and apoptosis through activating the Akt/HO-1 pathway, suggesting the protective effect of Oxy against H2O2-induced cataract. Hydrogen Peroxide 29-33 heme oxygenase 1 Homo sapiens 100-104 30709701-12 2019 In conclusion, Oxy protected H2O2-induced oxidative stress and apoptosis through activating the Akt/HO-1 pathway, suggesting the protective effect of Oxy against H2O2-induced cataract. Hydrogen Peroxide 162-166 heme oxygenase 1 Homo sapiens 100-104 30630055-10 2019 Pretreatment with deferoxamine, an inhibitor of heme synthesis, upregulated BACH1 protein levels and abolished the effect of NO-donors and S1PC on HMOX1 expression. Deferoxamine 18-30 heme oxygenase 1 Homo sapiens 147-152 29998398-10 2019 The cells were exposed to GAS (25 muM) for 4 h prior to the challenge with PQ at 100 muM for additional 24 h. The silencing of Nrf2 by siRNA or the inhibition of HO-1 by ZnPP IX suppressed the GAS-elicited cytoprotection. zinc protoporphyrin 170-177 heme oxygenase 1 Homo sapiens 162-166 30813369-0 2019 Anti-Inflammatory Effect of Sulforaphane on LPS-Activated Microglia Potentially through JNK/AP-1/NF-kappaB Inhibition and Nrf2/HO-1 Activation. sulforaphane 28-40 heme oxygenase 1 Homo sapiens 127-131 30746538-6 2019 PQ-induced DNA fragmentation in lymphocytes, reduction of oxidant scavenging capacity, expression of heme oxygenase 1 and inducible nitric oxide synthase in the lung, and elevation of serum transforming growth factor beta 1 were also inhibited. Paraquat 0-2 heme oxygenase 1 Homo sapiens 101-153 30783082-2 2019 Caffeic acid phenethyl ester (CAPE), which is a potent inducer of heme oxygenase 1 (HO1), is a central active component of propolis, and the application of propolis improves periodontal status in diabetic patients. caffeic acid phenethyl ester 0-28 heme oxygenase 1 Homo sapiens 66-82 30783082-2 2019 Caffeic acid phenethyl ester (CAPE), which is a potent inducer of heme oxygenase 1 (HO1), is a central active component of propolis, and the application of propolis improves periodontal status in diabetic patients. caffeic acid phenethyl ester 0-28 heme oxygenase 1 Homo sapiens 84-87 30783082-2 2019 Caffeic acid phenethyl ester (CAPE), which is a potent inducer of heme oxygenase 1 (HO1), is a central active component of propolis, and the application of propolis improves periodontal status in diabetic patients. caffeic acid phenethyl ester 30-34 heme oxygenase 1 Homo sapiens 66-82 30783082-2 2019 Caffeic acid phenethyl ester (CAPE), which is a potent inducer of heme oxygenase 1 (HO1), is a central active component of propolis, and the application of propolis improves periodontal status in diabetic patients. caffeic acid phenethyl ester 30-34 heme oxygenase 1 Homo sapiens 84-87 30605610-3 2019 As one of antioxidant defense systems, heme oxygenase-1 (HO-1) acts as an essential role in tumor development by offering antioxidant bilirubin to protect cancer cells under stress conditions. Bilirubin 134-143 heme oxygenase 1 Homo sapiens 39-55 30786886-9 2019 Mechanistically, DMO-CAP treatment induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK), JNK MAPK, and ERK MAPK, which led to the activation of Nrf2/heme oxygenase-1 (HO-1) pathway. Dimethadione 17-20 heme oxygenase 1 Homo sapiens 166-187 30786886-9 2019 Mechanistically, DMO-CAP treatment induced the phosphorylation of p38 mitogen-activated protein kinase (MAPK), JNK MAPK, and ERK MAPK, which led to the activation of Nrf2/heme oxygenase-1 (HO-1) pathway. Dimethadione 17-20 heme oxygenase 1 Homo sapiens 189-193 30759842-0 2019 Fourfold Filtered Statistical/Computational Approach for the Identification of Imidazole Compounds as HO-1 Inhibitors from Natural Products. imidazole 79-88 heme oxygenase 1 Homo sapiens 102-106 30759842-3 2019 Imidazole scaffold is normally present in most of the classical HO-1 inhibitors and seems indispensable to the inhibitory activity due to its strong interaction with the Fe(II) of the heme group. imidazole 0-9 heme oxygenase 1 Homo sapiens 64-68 30759842-3 2019 Imidazole scaffold is normally present in most of the classical HO-1 inhibitors and seems indispensable to the inhibitory activity due to its strong interaction with the Fe(II) of the heme group. ammonium ferrous sulfate 170-176 heme oxygenase 1 Homo sapiens 64-68 30759842-3 2019 Imidazole scaffold is normally present in most of the classical HO-1 inhibitors and seems indispensable to the inhibitory activity due to its strong interaction with the Fe(II) of the heme group. Heme 184-188 heme oxygenase 1 Homo sapiens 64-68 30759842-6 2019 Different imidazole-based compounds were suggested by our methodology to be potentially active in inhibiting the HO-1, and the results have been rationalized by the bioactivity of the filtered molecules reported in the literature. imidazole 10-19 heme oxygenase 1 Homo sapiens 113-117 30685970-8 2019 The results showed that Arg promoted the protein expression of Nrf2, up-regulated expression of the phase II metabolizing enzymes (NQO1 and HO-1), as well as antioxidative enzymes (GPx1, CAT, and SOD2) for alleviating oxidative injury and protected IOECs from LPS-induced apoptosis. Arginine 24-27 heme oxygenase 1 Homo sapiens 140-144 30605610-5 2019 To establish targeted anticancer therapy based on amplified oxidative stress, we developed molecularly engineered polymer, termed CZP, which incorporates ROS generating CA (cinnamaldehyde) and HO-1 inhibiting ZnPP (zinc protoporphyrin) in its backbone and could form stable micelles in aqueous solutions. zinc protoporphyrin 209-213 heme oxygenase 1 Homo sapiens 193-197 30605610-6 2019 CZP micelles not only elevated oxidative stress but also suppressed the expression of antioxidant HO-1, leading to apoptotic cell death. czp 0-3 heme oxygenase 1 Homo sapiens 98-102 30605610-3 2019 As one of antioxidant defense systems, heme oxygenase-1 (HO-1) acts as an essential role in tumor development by offering antioxidant bilirubin to protect cancer cells under stress conditions. Bilirubin 134-143 heme oxygenase 1 Homo sapiens 57-61 30605610-5 2019 To establish targeted anticancer therapy based on amplified oxidative stress, we developed molecularly engineered polymer, termed CZP, which incorporates ROS generating CA (cinnamaldehyde) and HO-1 inhibiting ZnPP (zinc protoporphyrin) in its backbone and could form stable micelles in aqueous solutions. czp 130-133 heme oxygenase 1 Homo sapiens 193-197 30804804-7 2019 The cytoprotective effects of HO-1 depend on several cellular mechanisms including the generation of bilirubin, an anti-oxidant molecule, from the degradation of heme; the induction of ferritin, a strong chelator of free iron; and the release of CO, that displays multiple anti-inflammatory and anti-apoptotic actions. Bilirubin 101-110 heme oxygenase 1 Homo sapiens 30-34 30804804-7 2019 The cytoprotective effects of HO-1 depend on several cellular mechanisms including the generation of bilirubin, an anti-oxidant molecule, from the degradation of heme; the induction of ferritin, a strong chelator of free iron; and the release of CO, that displays multiple anti-inflammatory and anti-apoptotic actions. Heme 162-166 heme oxygenase 1 Homo sapiens 30-34 30804804-7 2019 The cytoprotective effects of HO-1 depend on several cellular mechanisms including the generation of bilirubin, an anti-oxidant molecule, from the degradation of heme; the induction of ferritin, a strong chelator of free iron; and the release of CO, that displays multiple anti-inflammatory and anti-apoptotic actions. Iron 221-225 heme oxygenase 1 Homo sapiens 30-34 30324612-1 2019 OBJECTIVES: To evaluate the cytotoxic activity of atractylodin and its potential effects on heme oxygenase (HO)-1 production, STAT1/3 phosporylation and major NF-kappaB protein expression in the cholangiocarcinoma-associated cell line CL-6. atractylodin 50-62 heme oxygenase 1 Homo sapiens 92-113 30736789-14 2019 CONCLUSIONS: These results indicate that STL or STB may induce GSK3beta-dependent cyclin D1 degradation, and increase HO-1 expression through activating Nrf2 via ROS-dependent p38 activation, which resulted in the decrease of the viability in SW480 cells. ros 162-165 heme oxygenase 1 Homo sapiens 118-122 30840308-10 2019 Moreover, curcumin also decreased the expression of HIF-1alpha downstream genes, VEGF, HMOX1, ROS and PDGF. Curcumin 10-18 heme oxygenase 1 Homo sapiens 87-92 30535443-8 2019 Pretreatment with 3-morpholinosydnonimine hydrochloride, an inducer of the HO-1 expression, or overexpression of the HO-1 gene significantly delayed chondrocyte senescence. linsidomine 18-55 heme oxygenase 1 Homo sapiens 75-79 30736789-12 2019 In addition, STL or STB increased HO-1 expression, and the inhibition of HO-1 attenuated the induction of apoptosis by STL or STB. stb 20-23 heme oxygenase 1 Homo sapiens 34-38 30736789-13 2019 HO-1 expression by STL or STB resulted from Nrf2 activation through ROS-dependent p38 activation. ros 68-71 heme oxygenase 1 Homo sapiens 0-4 30815229-0 2019 Correction: 4-Hydroxyestradiol induces mammary epithelial cell transformation through Nrf2-mediated heme oxygenase-1 overexpression. 4-hydroxyestradiol 12-30 heme oxygenase 1 Homo sapiens 100-116 30476559-4 2019 Only in thyrocytes, H2O2 up-regulated GPx activity and expression of HO-1 mRNA. Hydrogen Peroxide 20-24 heme oxygenase 1 Homo sapiens 69-73 30718683-3 2019 Microarray analysis revealed that exposure of HUVECs to high MGO concentrations significantly changes gene expression, characterized by prominent down-regulation of cell cycle associated genes and up-regulation of heme oxygenase-1 (HO-1). Pyruvaldehyde 61-64 heme oxygenase 1 Homo sapiens 214-230 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Heme 148-152 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Heme 148-152 heme oxygenase 1 Homo sapiens 30-34 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Carbon Monoxide 165-180 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Carbon Monoxide 165-180 heme oxygenase 1 Homo sapiens 30-34 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Carbon Monoxide 182-184 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Carbon Monoxide 182-184 heme oxygenase 1 Homo sapiens 30-34 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Iron 187-191 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Iron 187-191 heme oxygenase 1 Homo sapiens 30-34 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Biliverdine 197-207 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Biliverdine 197-207 heme oxygenase 1 Homo sapiens 30-34 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Biliverdine 209-211 heme oxygenase 1 Homo sapiens 12-28 30580021-1 2019 BACKGROUND: Heme oxygenase-1 (HO-1), a cellular stress protein, serves a vital metabolic function as the rate-limiting enzyme in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin (BR). Biliverdine 209-211 heme oxygenase 1 Homo sapiens 30-34 30308911-11 2019 The higher content in PAHs measured in wood and charcoal PMs was likely the reason of the enhanced expression of metabolizing and oxidative stress-related enzymes, like CYP1B1 and HO-1, and the consequent increase in apoptotic cell death. Promethium 57-60 heme oxygenase 1 Homo sapiens 180-184 30628023-10 2019 High-glucose treatment for 24 h down-regulated the protein expression of redox-specific transcription factors Nrf-2, XBP-1 and NF-kappaB, and subsequently decreased the expression of HO-1, catalase, and SOD-2. Glucose 5-12 heme oxygenase 1 Homo sapiens 183-197 30529626-5 2019 Importantly, cordycepin treatment down-regulated the expression levels of Nuclear factor erythroid 2-related factor (Nrf2) and a series of downstream genes, such as heme oxygenase-1 (HO-1), to enhance ROS in breast cancer cells exposed to irradiation. Reactive Oxygen Species 201-204 heme oxygenase 1 Homo sapiens 165-181 30302660-8 2019 Since the prometastatic effects of bioactive phospholipids in vivo were mediated, at least in part, by downregulating HO-1 and iNOS expression in a p38 MAPK-dependent manner, we propose that inhibitors of p38 MAPK or stimulators of HO-1 activity will find application in inhibiting the spread of leukemic cells in response to bioactive phospholipids. Phospholipids 45-58 heme oxygenase 1 Homo sapiens 118-122 30302660-8 2019 Since the prometastatic effects of bioactive phospholipids in vivo were mediated, at least in part, by downregulating HO-1 and iNOS expression in a p38 MAPK-dependent manner, we propose that inhibitors of p38 MAPK or stimulators of HO-1 activity will find application in inhibiting the spread of leukemic cells in response to bioactive phospholipids. Phospholipids 45-58 heme oxygenase 1 Homo sapiens 232-236 30302660-0 2019 Bioactive Phospholipids Enhance Migration and Adhesion of Human Leukemic Cells by Inhibiting Heme Oxygenase 1 (HO-1) and Inducible Nitric Oxygenase Synthase (iNOS) in a p38 MAPK-Dependent Manner. Phospholipids 10-23 heme oxygenase 1 Homo sapiens 93-109 30302660-0 2019 Bioactive Phospholipids Enhance Migration and Adhesion of Human Leukemic Cells by Inhibiting Heme Oxygenase 1 (HO-1) and Inducible Nitric Oxygenase Synthase (iNOS) in a p38 MAPK-Dependent Manner. Phospholipids 10-23 heme oxygenase 1 Homo sapiens 111-115 30302660-5 2019 We also found that bioactive phospholipids enhanced cell migration and adhesion of leukemic cells by downregulating expression of HO-1 and iNOS in a p38 MAPK-dependent manner but did not affect cell proliferation. Phospholipids 29-42 heme oxygenase 1 Homo sapiens 130-134 30529626-5 2019 Importantly, cordycepin treatment down-regulated the expression levels of Nuclear factor erythroid 2-related factor (Nrf2) and a series of downstream genes, such as heme oxygenase-1 (HO-1), to enhance ROS in breast cancer cells exposed to irradiation. Reactive Oxygen Species 201-204 heme oxygenase 1 Homo sapiens 183-187 30529626-6 2019 Together, our observations demonstrate that cordycepin treatment sensitizes breast carcinoma cells toward irradiation via Nrf2/HO-1/ROS axis. Reactive Oxygen Species 132-135 heme oxygenase 1 Homo sapiens 127-131 30580124-0 2019 heme oxygenase-1 agonist CoPP suppresses influenza virus replication through IRF3-mediated generation of IFN-alpha/beta. COPP protocol 25-29 heme oxygenase 1 Homo sapiens 0-16 30580124-3 2019 In this study, we aim to confirm the antiviral effect of CoPP (Cobaltic Protoporphyrin IX Chloride), a potent HO-1 inducer on IAV infection and elucidate the possible mechanism of HO-1-mediated host innate immune responses. COPP protocol 57-61 heme oxygenase 1 Homo sapiens 110-114 30580124-3 2019 In this study, we aim to confirm the antiviral effect of CoPP (Cobaltic Protoporphyrin IX Chloride), a potent HO-1 inducer on IAV infection and elucidate the possible mechanism of HO-1-mediated host innate immune responses. COPP protocol 57-61 heme oxygenase 1 Homo sapiens 180-184 30580124-3 2019 In this study, we aim to confirm the antiviral effect of CoPP (Cobaltic Protoporphyrin IX Chloride), a potent HO-1 inducer on IAV infection and elucidate the possible mechanism of HO-1-mediated host innate immune responses. COBALTIC PROTOPORPHYRIN IX CHLORIDE 63-98 heme oxygenase 1 Homo sapiens 110-114 30837406-3 2019 The Nrf2/HO-1 axis counteracts oxidative stress injury by its resistance to inflammation, oxidation, mitochondrial damage and calcium influx, apoptosis, pyroptosis, ferroptosis and autophagy, which provides a theoretical basis for its therapeutic effect on various oxidative stress-relevant diseases in multiple organs (respiratory, cardiovascular, nervous, digestive, urinary and blood systems). Calcium 126-133 heme oxygenase 1 Homo sapiens 9-13 30696869-5 2019 Biochemical parameters were assayed and showed that the shift from normal glucose (NG; 5.5 mM) to high glucose (HG; 25 mM) promoted cell growth and induced oxidative/inflammatory stress and microglial activation, as evidenced by increased MTT reduction, elevated pro-inflammatory factor secretion (i.e., TNF-alpha and oxygen free radicals), and upregulated expression of stress/inflammatory proteins (i.e., HSP70, HO-1, iNOS, and COX-2). Glucose 103-110 heme oxygenase 1 Homo sapiens 414-418 30805085-10 2019 The mRNA and protein expression of Nrf2, HO-1, NQO1, and glutathione-S-transferase increased in the As + GSPE group compared with that in the As group (P < 0.05). Arsenic 100-102 heme oxygenase 1 Homo sapiens 41-45 30683864-7 2019 Collectively, our data indicate that HO-1, by detoxifying heme, blocks p16INK4a expression in macrophages, preventing DNA damage and cellular senescence. Heme 58-62 heme oxygenase 1 Homo sapiens 37-41 30678050-3 2019 Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. tin mesoporphyrin 0-17 heme oxygenase 1 Homo sapiens 45-61 30678050-3 2019 Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. tin mesoporphyrin 0-17 heme oxygenase 1 Homo sapiens 63-67 30678050-3 2019 Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. tin mesoporphyrin 19-23 heme oxygenase 1 Homo sapiens 45-61 30678050-3 2019 Tin mesoporphyrin (SnMP) is known to inhibit heme oxygenase-1 (HO-1), which has been shown to boost the activation and proliferation of human virus-specific T cells. tin mesoporphyrin 19-23 heme oxygenase 1 Homo sapiens 63-67 30678050-5 2019 HO-1 inhibition with SnMP increased WT1-specific T-cell frequencies in 13 (26%) of 50 healthy donors. tin mesoporphyrin 21-25 heme oxygenase 1 Homo sapiens 0-4 30678050-8 2019 In conclusion, pharmacological inhibition of HO-1 activity with SnMP results in more efficient generation of functionally active WT1-specific T cells. tin mesoporphyrin 64-68 heme oxygenase 1 Homo sapiens 45-49 31679277-0 2019 Nrf2/HO-1 Mediated Protective Activity of Genistein Against Doxorubicin-Induced Cardiac Toxicity. Genistein 42-51 heme oxygenase 1 Homo sapiens 5-9 31468438-6 2019 Moreover, APW increased the expressions of nuclear Nrf2 and cytosolic HO-1 in H2O2-treated hepatocytes. Hydrogen Peroxide 78-82 heme oxygenase 1 Homo sapiens 70-74 31468438-7 2019 Interestingly, the treatment of ZnPP, a HO-1 inhibitor abolished the cell viability and intracellular ROS generation induced by APW treatment. zinc protoporphyrin 32-36 heme oxygenase 1 Homo sapiens 40-44 31468438-7 2019 Interestingly, the treatment of ZnPP, a HO-1 inhibitor abolished the cell viability and intracellular ROS generation induced by APW treatment. Reactive Oxygen Species 102-105 heme oxygenase 1 Homo sapiens 40-44 30414976-10 2019 Overall, the work reveals that andrographolide through modulation of p53 and HNF4A, regulates miRNAs leading to upregulation of HO-1, glutathione and thioredoxin systems. andrographolide 31-46 heme oxygenase 1 Homo sapiens 128-132 31679277-14 2019 In conclusion, the potential antioxidant effect of genistein may be because of its modulatory effect on Nrf2/HO-1 signalling pathway and by this means exhibits cardioprotective effects from Dox-induced oxidative injury. Genistein 51-60 heme oxygenase 1 Homo sapiens 109-113 30621167-0 2019 Protective Effects and Mechanisms of N-Phenethyl Caffeamide from UVA-Induced Skin Damage in Human Epidermal Keratinocytes through Nrf2/HO-1 Regulation. N-phenethyl caffeamide 37-59 heme oxygenase 1 Homo sapiens 135-139 30529211-9 2019 Also, we demonstrated that BCP"s protective effects against LPS-induced oligodendrocytes toxicity were mediated via the CB2 receptor through different pathways including Nrf2/HO-1/anti-oxidant axis, and PPAR-gamma, at low (0.2 and 1 microM), and high (10-50 microM) concentrations, respectively. caryophyllene 27-30 heme oxygenase 1 Homo sapiens 175-179 31288720-4 2019 Recent studies revealed that heme oxygenase 1 (HO-1), an enzyme that participates in heme degradation, plays a critical role in the pathogenesis and may regulate autoimmunity. Heme 29-33 heme oxygenase 1 Homo sapiens 47-51 31032752-11 2019 Treatment with celastrol enhanced the expression of transcription factor Nrf2 and its targets, GCLM and HO-1. celastrol 15-24 heme oxygenase 1 Homo sapiens 104-108 30128961-7 2019 RESULTS: HO-1 mRNA, protein level, and HO-1 enzyme activity in renal tissue of rats with diabetic nephropathy were significantly increased after treatment with sitagliptin (P < 0.05). Sitagliptin Phosphate 160-171 heme oxygenase 1 Homo sapiens 39-43 30414976-2 2019 Our earlier studies have demonstrated the ability of andrographolide as well as andrographolide enriched extracts to activate Nrf2/HO-1 pathway through adenosine A2a receptor. andrographolide 53-68 heme oxygenase 1 Homo sapiens 131-135 30414976-2 2019 Our earlier studies have demonstrated the ability of andrographolide as well as andrographolide enriched extracts to activate Nrf2/HO-1 pathway through adenosine A2a receptor. andrographolide 80-95 heme oxygenase 1 Homo sapiens 131-135 30414976-7 2019 Downregulation of HNF4A by andrographolide led to decrease in miRNAs regulating Heme oxygenase-1 (miR-377) and glutathione cysteine ligase (miR-433). andrographolide 27-42 heme oxygenase 1 Homo sapiens 80-96 30256439-4 2019 The excess ROS generation will excite expression of heme oxygenase-1 and suppress CDA expression. Reactive Oxygen Species 11-14 heme oxygenase 1 Homo sapiens 52-68 30879014-0 2019 Vitamin D Ameliorates Angiotensin II-Induced Human Endothelial Progenitor Cell Injury via the PPAR-gamma/HO-1 Pathway. Vitamin D 0-9 heme oxygenase 1 Homo sapiens 105-109 30879014-9 2019 These findings also suggest that vitamin D protected EPCs from AngII-induced vascular injury via the activation of the PPAR-gamma/HO-1 signaling pathway. Vitamin D 33-42 heme oxygenase 1 Homo sapiens 130-134 31679277-0 2019 Nrf2/HO-1 Mediated Protective Activity of Genistein Against Doxorubicin-Induced Cardiac Toxicity. Doxorubicin 60-71 heme oxygenase 1 Homo sapiens 5-9 31679277-11 2019 Genistein enhanced the Nrf2 and HO-1 expression, which showed protection against oxidative insult induced by Dox. Genistein 0-9 heme oxygenase 1 Homo sapiens 32-36 31679277-11 2019 Genistein enhanced the Nrf2 and HO-1 expression, which showed protection against oxidative insult induced by Dox. 1-deoxysphinganine 109-112 heme oxygenase 1 Homo sapiens 32-36 30597920-5 2018 FBTA was capable of combating oxidative stress by augmenting messenger RNA (mRNA) and protein levels of both phase I and phase II detoxifying enzymes, especially heme oxygenase 1 (HO-1), by upregulating the nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated pathway in HaCaT cells via the phosphorylation of p38 and extracellular signal-regulated kinase (ERK). 2-(4-((2-Fluoroethyl)amino)phenyl)benzo[d]thiazol-6-ol 0-4 heme oxygenase 1 Homo sapiens 162-178 30444987-7 2019 SIGNIFICANCE: Pretreatment of crocetin and crocetin-loaded NPs provided pronounce protective effect against cyclosporine A-mediated toxicity in HEK-293 cells by nullifying the ROS formation and restored antioxidant network through inhibition of Nrf2 translocation and followed by expression of HO-1. crocetin 30-38 heme oxygenase 1 Homo sapiens 294-298 30444987-7 2019 SIGNIFICANCE: Pretreatment of crocetin and crocetin-loaded NPs provided pronounce protective effect against cyclosporine A-mediated toxicity in HEK-293 cells by nullifying the ROS formation and restored antioxidant network through inhibition of Nrf2 translocation and followed by expression of HO-1. crocetin 43-51 heme oxygenase 1 Homo sapiens 294-298 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Heme 37-41 heme oxygenase 1 Homo sapiens 127-143 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Heme 37-41 heme oxygenase 1 Homo sapiens 145-149 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 127-143 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 145-149 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Iron 73-77 heme oxygenase 1 Homo sapiens 127-143 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Iron 73-77 heme oxygenase 1 Homo sapiens 145-149 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Biliverdine 82-92 heme oxygenase 1 Homo sapiens 127-143 30009872-1 2019 Under stressful conditions, cellular heme catabolism to carbon monoxide, iron and biliverdin is mediated by the 32 kDa enzyme, heme oxygenase-1 (HO-1). Biliverdine 82-92 heme oxygenase 1 Homo sapiens 145-149 30009872-4 2019 By converting pro-oxidant heme to the antioxidants, biliverdin and bilirubin, HO-1/biliverdin reductase may help restore a more favorable tissue redox microenvironment. Heme 26-30 heme oxygenase 1 Homo sapiens 78-82 30009872-4 2019 By converting pro-oxidant heme to the antioxidants, biliverdin and bilirubin, HO-1/biliverdin reductase may help restore a more favorable tissue redox microenvironment. Bilirubin 67-76 heme oxygenase 1 Homo sapiens 78-82 30009872-7 2019 A comprehensive model of astroglial stress is presented wherein sustained Hmox1 induction promotes oxidative mitochondrial membrane damage, iron sequestration and mitophagy (macroautophagy). Iron 140-144 heme oxygenase 1 Homo sapiens 74-79 30346043-0 2019 Berberine ameliorates lipopolysaccharide-induced acute lung injury via the PERK-mediated Nrf2/HO-1 signaling axis. Berberine 0-9 heme oxygenase 1 Homo sapiens 94-98 30346043-12 2019 Therefore, berberine inhibits LPS-induced ALI through the PERK-mediated Nrf2/HO-1 signaling axis. Berberine 11-20 heme oxygenase 1 Homo sapiens 77-81 30393147-10 2019 Furthermore, pioglitazone increased the expression of PPARgamma target genes such as renal and hepatic PPARgamma1 (Pparg1), hepatic hemoxygenase-1 (Hmox1), and hepatic thioredoxin (TRx). Pioglitazone 13-25 heme oxygenase 1 Homo sapiens 132-146 30393147-10 2019 Furthermore, pioglitazone increased the expression of PPARgamma target genes such as renal and hepatic PPARgamma1 (Pparg1), hepatic hemoxygenase-1 (Hmox1), and hepatic thioredoxin (TRx). Pioglitazone 13-25 heme oxygenase 1 Homo sapiens 148-153 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Carbon Monoxide 90-105 heme oxygenase 1 Homo sapiens 0-16 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Carbon Monoxide 90-105 heme oxygenase 1 Homo sapiens 18-22 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Iron 112-116 heme oxygenase 1 Homo sapiens 0-16 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Iron 112-116 heme oxygenase 1 Homo sapiens 18-22 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Biliverdine 122-132 heme oxygenase 1 Homo sapiens 0-16 30213580-3 2019 Heme oxygenase-1 (HO-1) plays a crucial role in heme degradation and in this way releases carbon monoxide, free iron, and biliverdin. Biliverdine 122-132 heme oxygenase 1 Homo sapiens 18-22 30213580-5 2019 Consequently, biological actions of HO-1 are not limited to degradation of a toxic heme released from hemoproteins, but also provide an adaptive cellular response against chronic inflammation and oxidative injury. Heme 83-87 heme oxygenase 1 Homo sapiens 36-40 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 32-55 heme oxygenase 1 Homo sapiens 108-112 30583467-1 2018 Heme oxygenase (HO)-1 is known to metabolize heme into biliverdin/bilirubin, carbon monoxide, and ferrous iron, and it has been suggested to demonstrate cytoprotective effects against various stress-related conditions. Heme 45-49 heme oxygenase 1 Homo sapiens 0-21 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 32-55 heme oxygenase 1 Homo sapiens 171-175 30583467-1 2018 Heme oxygenase (HO)-1 is known to metabolize heme into biliverdin/bilirubin, carbon monoxide, and ferrous iron, and it has been suggested to demonstrate cytoprotective effects against various stress-related conditions. Biliverdine 55-65 heme oxygenase 1 Homo sapiens 0-21 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 57-60 heme oxygenase 1 Homo sapiens 108-112 30583467-1 2018 Heme oxygenase (HO)-1 is known to metabolize heme into biliverdin/bilirubin, carbon monoxide, and ferrous iron, and it has been suggested to demonstrate cytoprotective effects against various stress-related conditions. Bilirubin 66-75 heme oxygenase 1 Homo sapiens 0-21 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 57-60 heme oxygenase 1 Homo sapiens 171-175 30583467-1 2018 Heme oxygenase (HO)-1 is known to metabolize heme into biliverdin/bilirubin, carbon monoxide, and ferrous iron, and it has been suggested to demonstrate cytoprotective effects against various stress-related conditions. Carbon Monoxide 77-92 heme oxygenase 1 Homo sapiens 0-21 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Iron 142-146 heme oxygenase 1 Homo sapiens 108-112 30583467-1 2018 Heme oxygenase (HO)-1 is known to metabolize heme into biliverdin/bilirubin, carbon monoxide, and ferrous iron, and it has been suggested to demonstrate cytoprotective effects against various stress-related conditions. Iron 106-110 heme oxygenase 1 Homo sapiens 0-21 30583467-5 2018 The critical role of HO-1 in heme metabolism makes it an important candidate to mediate protective or detrimental effects via ferroptosis induction. Heme 29-33 heme oxygenase 1 Homo sapiens 21-25 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Iron 142-146 heme oxygenase 1 Homo sapiens 171-175 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Iron 23-27 heme oxygenase 1 Homo sapiens 108-112 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 151-154 heme oxygenase 1 Homo sapiens 108-112 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Iron 23-27 heme oxygenase 1 Homo sapiens 171-175 30583467-7 2018 The amount of cellular iron and reactive oxygen species (ROS) is the determinative momentum for the role of HO-1, in which excessive cellular iron and ROS tend to enforce HO-1 from a protective role to a perpetrator. Reactive Oxygen Species 151-154 heme oxygenase 1 Homo sapiens 171-175 30577437-1 2018 Heme oxygenase-1 (HO-1), a rate-limiting enzyme involved in the degradation of heme, is induced in response to a wide range of stress conditions. Heme 79-83 heme oxygenase 1 Homo sapiens 0-16 30619356-0 2018 Heme Drives Susceptibility of Glomerular Endothelium to Complement Overactivation Due to Inefficient Upregulation of Heme Oxygenase-1. Heme 0-4 heme oxygenase 1 Homo sapiens 117-133 30619356-8 2018 Only HUVEC (Human Umbilical Vein EC) developed adaptation to heme, which was lost after inhibition of HO-1 activity. Heme 61-65 heme oxygenase 1 Homo sapiens 102-106 30577437-1 2018 Heme oxygenase-1 (HO-1), a rate-limiting enzyme involved in the degradation of heme, is induced in response to a wide range of stress conditions. Heme 79-83 heme oxygenase 1 Homo sapiens 18-22 30321531-0 2018 A new chalcone derivative, 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one), inhibits phorbol ester-induced metastatic activity of colorectal cancer cells through upregulation of heme oxygenase-1. Chalcone 6-14 heme oxygenase 1 Homo sapiens 192-208 30647815-8 2018 Results: We found increased levels of ROS and increased expression of ROS-producing enzymes (i.e., p47phox, xanthine oxidase) and decreased antioxidant defense mechanisms (mitochondrial aldehyde dehydrogenase, heme oxygenase-1, and eNOS) in line with elevated inflammatory markers (vascular cell adhesion molecule-1) in the right atrial myocardial tissue and by trend also in serum (sVCAM-1 and CCL5/RANTES). Reactive Oxygen Species 38-41 heme oxygenase 1 Homo sapiens 210-226 30142309-10 2018 We used zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, to elucidate the relationship between HO-1 expression and cell death in vitro in a hemin injury model. zinc protoporphyrin 8-27 heme oxygenase 1 Homo sapiens 56-60 30142309-10 2018 We used zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, to elucidate the relationship between HO-1 expression and cell death in vitro in a hemin injury model. zinc protoporphyrin 8-27 heme oxygenase 1 Homo sapiens 111-115 30142309-10 2018 We used zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, to elucidate the relationship between HO-1 expression and cell death in vitro in a hemin injury model. zinc protoporphyrin 29-33 heme oxygenase 1 Homo sapiens 56-60 30142309-10 2018 We used zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, to elucidate the relationship between HO-1 expression and cell death in vitro in a hemin injury model. zinc protoporphyrin 29-33 heme oxygenase 1 Homo sapiens 111-115 30321531-0 2018 A new chalcone derivative, 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one), inhibits phorbol ester-induced metastatic activity of colorectal cancer cells through upregulation of heme oxygenase-1. 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one 27-87 heme oxygenase 1 Homo sapiens 192-208 30321531-0 2018 A new chalcone derivative, 3-phenyl-1-(2,4,6-tris(methoxymethoxy)phenyl)prop-2-yn-1-one), inhibits phorbol ester-induced metastatic activity of colorectal cancer cells through upregulation of heme oxygenase-1. Phorbol Esters 99-112 heme oxygenase 1 Homo sapiens 192-208 30321531-11 2018 In conclusion, KB-34 inhibits the TPA-stimulated metastatic potential of HT-29 cells by induction of HO-1 and may be a promising anti-cancer agent in chemotherapeutic strategies for CRC. kb-34 15-20 heme oxygenase 1 Homo sapiens 101-105 30321531-11 2018 In conclusion, KB-34 inhibits the TPA-stimulated metastatic potential of HT-29 cells by induction of HO-1 and may be a promising anti-cancer agent in chemotherapeutic strategies for CRC. Tetradecanoylphorbol Acetate 34-37 heme oxygenase 1 Homo sapiens 101-105 30563120-10 2018 Our previous studies have documented that bisdemethoxycurcumin (BDMC) induces apoptosis by regulating heme oxygenase (HO)-1 protein expression in activated HSCs. bisdemethoxycurcumin 42-62 heme oxygenase 1 Homo sapiens 102-123 28793787-7 2018 Constitutive NRF2 activation and subsequent deregulation of iron metabolism have been implicated in cancer development: NRF2-mediated upregulation of the iron storage protein ferritin or heme oxygenase 1 can lead to enhanced proliferation and therapy resistance. Iron 60-64 heme oxygenase 1 Homo sapiens 187-203 30563120-10 2018 Our previous studies have documented that bisdemethoxycurcumin (BDMC) induces apoptosis by regulating heme oxygenase (HO)-1 protein expression in activated HSCs. bisdemethoxycurcumin 64-68 heme oxygenase 1 Homo sapiens 102-123 30563120-12 2018 We observed that D2O cotreatment with BDMC significantly decreased cell proliferation compared to treatment with D2O alone, and this effect was accompanied by downregulation of HO-1 and an increase in p53 levels. Deuterium Oxide 17-20 heme oxygenase 1 Homo sapiens 177-181 30563120-12 2018 We observed that D2O cotreatment with BDMC significantly decreased cell proliferation compared to treatment with D2O alone, and this effect was accompanied by downregulation of HO-1 and an increase in p53 levels. bisdemethoxycurcumin 38-42 heme oxygenase 1 Homo sapiens 177-181 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 0-11 heme oxygenase 1 Homo sapiens 157-173 30372823-0 2018 Brahma-related gene 1 ameliorates the neuronal apoptosis and oxidative stress induced by oxygen-glucose deprivation/reoxygenation through activation of Nrf2/HO-1 signaling. oxygen-glucose 89-103 heme oxygenase 1 Homo sapiens 157-161 30548662-0 2018 SIRT3 inhibited the formation of calcium oxalate-induced kidney stones through regulating NRF2/HO-1 signaling pathway. Calcium Oxalate 33-48 heme oxygenase 1 Homo sapiens 95-99 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 0-11 heme oxygenase 1 Homo sapiens 175-179 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 0-3 heme oxygenase 1 Homo sapiens 157-173 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 0-3 heme oxygenase 1 Homo sapiens 175-179 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 13-16 heme oxygenase 1 Homo sapiens 157-173 30159756-1 2018 Doxorubicin (Dox)-induced cardiotoxicity, a limiting factor in the use of Dox to treat cancer, can be mitigated by the mitogenic factor FGF2 in vitro, via a heme oxygenase 1 (HO-1)-dependent pathway. Doxorubicin 13-16 heme oxygenase 1 Homo sapiens 175-179 30243774-6 2018 We observe that BBC results in a higher phosphorylation of PKCalpha/beta that is involved in the activation of erythroid 2-related factor 2 (Nrf2) phosphorylation to favor Nrf2 translocation to nucleus at 24 and 48 h. In addition, the results show that BBC also up-regulates both antiviral responses, heme oxygenase-1 (HO-1) expression and cellular IFN response. Benomyl 16-19 heme oxygenase 1 Homo sapiens 301-317 30243774-6 2018 We observe that BBC results in a higher phosphorylation of PKCalpha/beta that is involved in the activation of erythroid 2-related factor 2 (Nrf2) phosphorylation to favor Nrf2 translocation to nucleus at 24 and 48 h. In addition, the results show that BBC also up-regulates both antiviral responses, heme oxygenase-1 (HO-1) expression and cellular IFN response. Benomyl 253-256 heme oxygenase 1 Homo sapiens 301-317 30280183-5 2018 The function of protein kinase B (AKT) and heme oxygenase-1 (HO-1) activation on luteolin mediated I/R injury protection was assessed by administration of phosphoinositide-3-kinase/AKT inhibitor LY294002 and HO-1 inhibitor ZNPP. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 195-203 heme oxygenase 1 Homo sapiens 61-65 30238818-0 2018 Role of heme oxygenase-1 and its reaction product, carbon monoxide, in manifestation of breast cancer stem cell-like properties: Notch-1 as a putative target. Carbon Monoxide 51-66 heme oxygenase 1 Homo sapiens 8-24 30238818-5 2018 Carbon monoxide (CO) is endogenously generated as a consequence of degradation of heme by HO-1. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 90-94 30238818-5 2018 Carbon monoxide (CO) is endogenously generated as a consequence of degradation of heme by HO-1. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 90-94 30272282-6 2018 Furthermore, Sch B enhanced the expression of key antioxidant enzymes, including catalase, heme oxygenase-1, glutathione peroxidase, and superoxide dismutase, and further engaged the nuclear factor-erythroid 2-related factor 2 (Nrf2) signaling pathway by modulating its phosphorylation through activating multiple upstream kinases, including protein kinase B, adenosine monophosphate-activated protein kinase and mitogen-activated protein kinases (MAPKs). schizandrin B 13-18 heme oxygenase 1 Homo sapiens 91-107 30575934-0 2018 MicroRNA-29a enhances autophagy in podocytes as a protective mechanism against high glucose-induced apoptosis by targeting heme oxygenase-1. Glucose 84-91 heme oxygenase 1 Homo sapiens 123-139 32186127-7 2018 EXS-induced HO-1 expression was significantly decreased in SnPP (HO-1 inhibitor)- and HO-1 siRNA-treated cells, whereas an increase was found in cobalt protoporphyrin IX (CoPP) (HO-1 inducer)-treated cells. S-Nitroso-N-propionyl-D,L-penicillamine 59-63 heme oxygenase 1 Homo sapiens 12-16 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Heme 78-82 heme oxygenase 1 Homo sapiens 57-62 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Heme 78-82 heme oxygenase 1 Homo sapiens 96-100 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Iron 134-138 heme oxygenase 1 Homo sapiens 57-62 30272289-11 2018 The results revealed that treatment with TCSGs markedly induced Nrf2 nuclear translocation and upregulated the expression of heme oxygenase-1 (HO-1) in the L02 cells damaged by H2O2. tcsgs 41-46 heme oxygenase 1 Homo sapiens 125-141 30272289-11 2018 The results revealed that treatment with TCSGs markedly induced Nrf2 nuclear translocation and upregulated the expression of heme oxygenase-1 (HO-1) in the L02 cells damaged by H2O2. Hydrogen Peroxide 177-181 heme oxygenase 1 Homo sapiens 125-141 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Iron 134-138 heme oxygenase 1 Homo sapiens 78-94 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Iron 134-138 heme oxygenase 1 Homo sapiens 96-100 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Carbon Monoxide 140-155 heme oxygenase 1 Homo sapiens 57-62 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Carbon Monoxide 140-155 heme oxygenase 1 Homo sapiens 78-94 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Carbon Monoxide 140-155 heme oxygenase 1 Homo sapiens 96-100 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Biliverdine 161-171 heme oxygenase 1 Homo sapiens 57-62 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Biliverdine 161-171 heme oxygenase 1 Homo sapiens 78-94 30145824-2 2018 Bach1 is a mammalian transcription factor that represses Hmox1, which encodes heme oxygenase-1 (HO-1) that can degrade heme into free iron, carbon monoxide, and biliverdin, to play an important role in antioxidant, anti-inflammatory, and antiapoptotic activities. Biliverdine 161-171 heme oxygenase 1 Homo sapiens 96-100 32186127-7 2018 EXS-induced HO-1 expression was significantly decreased in SnPP (HO-1 inhibitor)- and HO-1 siRNA-treated cells, whereas an increase was found in cobalt protoporphyrin IX (CoPP) (HO-1 inducer)-treated cells. S-Nitroso-N-propionyl-D,L-penicillamine 59-63 heme oxygenase 1 Homo sapiens 65-69 30143976-0 2018 Curcumin confers hepatoprotection against AFB1-induced toxicity via activating autophagy and ameliorating inflammation involving Nrf2/HO-1 signaling pathway. Curcumin 0-8 heme oxygenase 1 Homo sapiens 134-138 32186127-7 2018 EXS-induced HO-1 expression was significantly decreased in SnPP (HO-1 inhibitor)- and HO-1 siRNA-treated cells, whereas an increase was found in cobalt protoporphyrin IX (CoPP) (HO-1 inducer)-treated cells. S-Nitroso-N-propionyl-D,L-penicillamine 59-63 heme oxygenase 1 Homo sapiens 65-69 30143976-0 2018 Curcumin confers hepatoprotection against AFB1-induced toxicity via activating autophagy and ameliorating inflammation involving Nrf2/HO-1 signaling pathway. Aflatoxin B1 42-46 heme oxygenase 1 Homo sapiens 134-138 32186127-7 2018 EXS-induced HO-1 expression was significantly decreased in SnPP (HO-1 inhibitor)- and HO-1 siRNA-treated cells, whereas an increase was found in cobalt protoporphyrin IX (CoPP) (HO-1 inducer)-treated cells. S-Nitroso-N-propionyl-D,L-penicillamine 59-63 heme oxygenase 1 Homo sapiens 65-69 30143976-9 2018 Moreover, curcumin treatment significantly (p < 0.05) elevated AFB1-induced decrease in Nrf2 and HO-1 mRNA and protein expression level. Curcumin 10-18 heme oxygenase 1 Homo sapiens 100-104 32186127-8 2018 In addition, pretreatment with EXS increased HO-1 and Nrf-2 expression under TNF-alpha stimulation with or without N-acetyl-L-cysteine. (2R)-2,4-dihydroxy-N-[2-(7-hydroxy-1H-benzimidazol-2-yl)ethyl]-3,3-dimethylbutanamide 31-34 heme oxygenase 1 Homo sapiens 45-49 30382449-4 2018 In addition, we revealed that PFOS exposure caused marked activation of Nrf2 pathway and the expression of Nrf2 transcription target heme oxygenase-1. perfluorooctane sulfonic acid 30-34 heme oxygenase 1 Homo sapiens 133-149 30439604-0 2018 Celastrol antagonizes high glucose-evoked podocyte injury, inflammation and insulin resistance by restoring the HO-1-mediated autophagy pathway. celastrol 0-9 heme oxygenase 1 Homo sapiens 112-116 30439604-11 2018 Importantly, celastrol enhanced heme oxygenase-1 (HO-1) expression in HG-stimulated podocytes. celastrol 13-22 heme oxygenase 1 Homo sapiens 32-48 30439604-11 2018 Importantly, celastrol enhanced heme oxygenase-1 (HO-1) expression in HG-stimulated podocytes. celastrol 13-22 heme oxygenase 1 Homo sapiens 50-54 30439604-12 2018 Notably, HO-1 cessation depressed autophagy pathway activation and subsequently blunted beneficial effects of celastrol on HG-exposed podocytes. celastrol 110-119 heme oxygenase 1 Homo sapiens 9-13 30439604-13 2018 These finding suggest that celastrol may protect against HG-induced podocyte injury, inflammation and insulin resistance by restoring HO-1-mediated autophagy pathway, implying a promising therapeutic strategy against DN. celastrol 27-36 heme oxygenase 1 Homo sapiens 134-138 30515391-8 2018 Our results were further confirmed by specific HO-1 gene knockdown studies which clearly demonstrated that HO-1 induction indeed played a key role in SAC mediated inhibition of apoptosis and ROS production in HepG2 cells, thus suggesting a hepatoprotective role of SAC in combating oxidative stress mediated liver diseases. Reactive Oxygen Species 191-194 heme oxygenase 1 Homo sapiens 107-111 29782798-5 2018 Moreover, in primary pan-microglial marker (CD11b)-positive cells, the major producers of inflammatory mediators in the brain, DA and its derivatives significantly diminish inflammation and oxidative stress triggered by lipopolysaccharides and Abeta through the reduced induction of inflammatory mediators as well as upregulated expression of heme oxygenase-1, the enzyme responsible for production of antioxidants. Dopamine 127-129 heme oxygenase 1 Homo sapiens 343-359 30248308-3 2018 In this paper, we investigated the role of HO-1 activation in NTP-induced apoptosis in A549 cells. ntp 62-65 heme oxygenase 1 Homo sapiens 43-47 30248308-5 2018 NTP exposure induced HO-1 expression in a dose- and time-dependent manner via activating the translocation of Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) from cytoplasm to nucleus. ntp 0-3 heme oxygenase 1 Homo sapiens 21-25 30248308-6 2018 Furthermore, inhibiting HO-1 activation with its specific inhibitor, ZnPP, increased "killing" effect of NTP. zinc protoporphyrin 69-73 heme oxygenase 1 Homo sapiens 24-28 30248308-6 2018 Furthermore, inhibiting HO-1 activation with its specific inhibitor, ZnPP, increased "killing" effect of NTP. ntp 105-108 heme oxygenase 1 Homo sapiens 24-28 30248308-7 2018 Knocking down HO-1 or Nrf2 with the special siRNA also led to elevated ROS level and enhanced NTP-induced cell death. Reactive Oxygen Species 71-74 heme oxygenase 1 Homo sapiens 14-18 30248308-7 2018 Knocking down HO-1 or Nrf2 with the special siRNA also led to elevated ROS level and enhanced NTP-induced cell death. ntp 94-97 heme oxygenase 1 Homo sapiens 14-18 30248308-8 2018 In addition, the c-JUN N-terminal kinase (JNK) signaling pathway was shown to be involved in NTP-induced HO-1 expression. ntp 93-96 heme oxygenase 1 Homo sapiens 105-109 30248308-10 2018 These findings revealed that HO-1 could be considered as a potential target to improve the effect of NTP in cancer therapy. ntp 101-104 heme oxygenase 1 Homo sapiens 29-33 30428359-4 2018 HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Oxygen 107-113 heme oxygenase 1 Homo sapiens 0-4 30419705-8 2018 In all groups ROS levels were detected after exposed to ZnO-NPs for 4 hours, the protein expression of HO-1 was detected after exposed to ZnO-NPs for 24 hours. Zinc Oxide 138-141 heme oxygenase 1 Homo sapiens 103-107 30419705-10 2018 HO-1 expression in ZnPPIx pretreatment group decreased compared with ZnO-NPs group (1.05+-0.05 vs. 1.12+-0.01, P<0.05), meanwhile ROS level enhanced (62 683.95+-2 589.59 vs. 53 654.53+-2 229.01, P<0.05). zinc protoporphyrin 19-25 heme oxygenase 1 Homo sapiens 0-4 30419705-10 2018 HO-1 expression in ZnPPIx pretreatment group decreased compared with ZnO-NPs group (1.05+-0.05 vs. 1.12+-0.01, P<0.05), meanwhile ROS level enhanced (62 683.95+-2 589.59 vs. 53 654.53+-2 229.01, P<0.05). Zinc Oxide 69-72 heme oxygenase 1 Homo sapiens 0-4 30419705-10 2018 HO-1 expression in ZnPPIx pretreatment group decreased compared with ZnO-NPs group (1.05+-0.05 vs. 1.12+-0.01, P<0.05), meanwhile ROS level enhanced (62 683.95+-2 589.59 vs. 53 654.53+-2 229.01, P<0.05). Reactive Oxygen Species 133-136 heme oxygenase 1 Homo sapiens 0-4 30419705-11 2018 However, CoPPIx pretreatment had higher HO-1 level and lower level of ROS compared with ZnO-NPs group (HO-1: 1.74+-0.11 vs. 0.22+-0.03, P<0.05; ROS: 32 845.04+-993.48 vs. 53 654.53+-2 229.01, P<0.05). coppix 9-15 heme oxygenase 1 Homo sapiens 40-44 30419705-11 2018 However, CoPPIx pretreatment had higher HO-1 level and lower level of ROS compared with ZnO-NPs group (HO-1: 1.74+-0.11 vs. 0.22+-0.03, P<0.05; ROS: 32 845.04+-993.48 vs. 53 654.53+-2 229.01, P<0.05). coppix 9-15 heme oxygenase 1 Homo sapiens 103-107 30419705-13 2018 HO-1 regulated ZnO-NPs-induced oxidative stress. Zinc Oxide 15-18 heme oxygenase 1 Homo sapiens 0-4 30354231-0 2018 Intravenous Heme Arginate Induces HO-1 (Heme Oxygenase-1) in the Human Heart. heme arginate 12-25 heme oxygenase 1 Homo sapiens 34-38 30354231-0 2018 Intravenous Heme Arginate Induces HO-1 (Heme Oxygenase-1) in the Human Heart. heme arginate 12-25 heme oxygenase 1 Homo sapiens 40-56 30354231-3 2018 The current trial was designed to assess the pharmacological tissue induction of HO-1 in the human heart with heme arginate in vivo. heme arginate 110-123 heme oxygenase 1 Homo sapiens 81-85 30354231-11 2018 Surgery and heme arginate infusion significantly increased HO-1 mRNA concentration in peripheral blood mononuclear cells ( P<0.001). Heme 12-16 heme oxygenase 1 Homo sapiens 59-63 30354231-11 2018 Surgery and heme arginate infusion significantly increased HO-1 mRNA concentration in peripheral blood mononuclear cells ( P<0.001). L-argininate 17-25 heme oxygenase 1 Homo sapiens 59-63 30354231-14 2018 Conclusions- Myocardial HO-1 mRNA and protein can be dose-dependently induced by heme arginate. heme arginate 81-94 heme oxygenase 1 Homo sapiens 24-28 30515391-0 2018 S-Allyl Cysteine Alleviates Hydrogen Peroxide Induced Oxidative Injury and Apoptosis through Upregulation of Akt/Nrf-2/HO-1 Signaling Pathway in HepG2 Cells. S-allylcysteine 0-16 heme oxygenase 1 Homo sapiens 119-123 30268863-13 2018 p38MAPK, JNK and HO-1 were activated after PDT treatment and the activation were reversed by adding ROS scavenger NAC. Reactive Oxygen Species 100-103 heme oxygenase 1 Homo sapiens 17-21 30428359-4 2018 HO-1 mitigates TB pathophysiology by diminishing myeloid cell-mediated oxidative damage caused by reactive oxygen and/or nitrogen intermediates, which control granulocytic karyorrhexis to generate a zonal HO-1 response. Nitrogen 121-129 heme oxygenase 1 Homo sapiens 0-4 30419705-0 2018 [The role of heme oxygenase-1 on oxidative stress injury induced by zinc oxide nanoparticles in human umbilical vein endothelial cells line EA.hy926 cells]. Zinc Oxide 68-78 heme oxygenase 1 Homo sapiens 13-29 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Reactive Oxygen Species 72-95 heme oxygenase 1 Homo sapiens 36-52 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Reactive Oxygen Species 72-95 heme oxygenase 1 Homo sapiens 54-58 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Reactive Oxygen Species 97-100 heme oxygenase 1 Homo sapiens 36-52 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Reactive Oxygen Species 97-100 heme oxygenase 1 Homo sapiens 54-58 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Zinc Oxide 113-123 heme oxygenase 1 Homo sapiens 36-52 30419705-1 2018 Objective: To explore the effect of heme oxygenase-1 (HO-1) on level of reactive oxygen species (ROS) induced by zinc oxide nanoparticles (ZnO-NPs) in Human umbilical vein endothelial cells line EA.hy926. Zinc Oxide 113-123 heme oxygenase 1 Homo sapiens 54-58 30419705-3 2018 The ROS level, reflected by mean fluorescence intensity (MFI), was examined by flow cytometer after 4 hours exposure, the protein expression of HO-1 which was determined by Western Blot after exposed to ZnO-NPs for 24 hours. Zinc Oxide 203-206 heme oxygenase 1 Homo sapiens 144-148 30419705-6 2018 15 mg/L ZnO-NPs was chosen to conduct the experiment of HO-1 activation and inhibition. Zinc Oxide 8-11 heme oxygenase 1 Homo sapiens 56-60 30419705-8 2018 In all groups ROS levels were detected after exposed to ZnO-NPs for 4 hours, the protein expression of HO-1 was detected after exposed to ZnO-NPs for 24 hours. Zinc Oxide 56-59 heme oxygenase 1 Homo sapiens 103-107 30257325-7 2018 Furthermore, eriodictyol pretreatment inhibited IkappaBalpha degradation and the level of p-p65, and enhanced the up-regulation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase 1 (HO-1) in IL-1beta-stimulated chondrocytes. eriodictyol 13-24 heme oxygenase 1 Homo sapiens 186-202 30257325-7 2018 Furthermore, eriodictyol pretreatment inhibited IkappaBalpha degradation and the level of p-p65, and enhanced the up-regulation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) and heme oxygenase 1 (HO-1) in IL-1beta-stimulated chondrocytes. eriodictyol 13-24 heme oxygenase 1 Homo sapiens 204-208 30216645-0 2018 Heme oxygenase-1 induction mediates chemoresistance of breast cancer cells to pharmorubicin by promoting autophagy via PI3K/Akt pathway. Epirubicin 78-91 heme oxygenase 1 Homo sapiens 0-16 30216645-8 2018 RESULTS: After being treated with pharmorubicin, the expression of HO-1 and autophagy related proteins was significantly enhanced, but the cell survival ratio in the two cell lines decreased. Epirubicin 34-47 heme oxygenase 1 Homo sapiens 67-71 30216645-10 2018 When pharmorubicin-resistant cells were transfected with si-HO-1, the cell survival decreased and the protein expression of HO-1, autophagic proteins (LC3-II/LC3-I and Beclin-1) as well as autophagy were all down-regulated, while in pharmorubicin-resistant cells transfected with pcDNA3.1-HO-1, the results were reverse. Epirubicin 5-18 heme oxygenase 1 Homo sapiens 60-64 30216645-10 2018 When pharmorubicin-resistant cells were transfected with si-HO-1, the cell survival decreased and the protein expression of HO-1, autophagic proteins (LC3-II/LC3-I and Beclin-1) as well as autophagy were all down-regulated, while in pharmorubicin-resistant cells transfected with pcDNA3.1-HO-1, the results were reverse. Epirubicin 5-18 heme oxygenase 1 Homo sapiens 124-128 30216645-10 2018 When pharmorubicin-resistant cells were transfected with si-HO-1, the cell survival decreased and the protein expression of HO-1, autophagic proteins (LC3-II/LC3-I and Beclin-1) as well as autophagy were all down-regulated, while in pharmorubicin-resistant cells transfected with pcDNA3.1-HO-1, the results were reverse. Epirubicin 5-18 heme oxygenase 1 Homo sapiens 124-128 30216645-10 2018 When pharmorubicin-resistant cells were transfected with si-HO-1, the cell survival decreased and the protein expression of HO-1, autophagic proteins (LC3-II/LC3-I and Beclin-1) as well as autophagy were all down-regulated, while in pharmorubicin-resistant cells transfected with pcDNA3.1-HO-1, the results were reverse. Epirubicin 233-246 heme oxygenase 1 Homo sapiens 60-64 30216645-12 2018 CONCLUSION: It was proved that HO-1 induction mediated chemoresistance of pharmorubicin in breast cancer cells by promoting autophagy via PI3K/Akt pathway. Epirubicin 74-87 heme oxygenase 1 Homo sapiens 31-35 29707997-10 2018 Linear regression analysis demonstrated that HO-1 was the independent predictor of AKI demonstrated by elevated creatinine among GI and GII. Creatinine 112-122 heme oxygenase 1 Homo sapiens 45-49 29707997-12 2018 HO-1 is a promising target for prevention and/or treatment of cisplatin-induced nephrotoxicity. Cisplatin 62-71 heme oxygenase 1 Homo sapiens 0-4 30397576-2 2018 Here, we tested the ability of different isoxazoline-based electrophiles to up-regulate Nrf2/HO-1. Isoxazoline 41-52 heme oxygenase 1 Homo sapiens 93-97 30367621-0 2018 Sorafenib and docosahexaenoic acid act in synergy to suppress cancer cell viability: a role of heme oxygenase 1. Sorafenib 0-9 heme oxygenase 1 Homo sapiens 95-111 30367621-0 2018 Sorafenib and docosahexaenoic acid act in synergy to suppress cancer cell viability: a role of heme oxygenase 1. Docosahexaenoic Acids 14-34 heme oxygenase 1 Homo sapiens 95-111 30367621-3 2018 We recently reported that DHA induces HO-1 gene transcription in human cancer cells by augmenting the degradation of Bach1 protein, which functions as a negative regulator of HO-1. Docosahexaenoic Acids 26-29 heme oxygenase 1 Homo sapiens 38-42 30367621-3 2018 We recently reported that DHA induces HO-1 gene transcription in human cancer cells by augmenting the degradation of Bach1 protein, which functions as a negative regulator of HO-1. Docosahexaenoic Acids 26-29 heme oxygenase 1 Homo sapiens 175-179 30206627-7 2018 Moreover, the combination treatments reduced reactive oxygen species in cells and increased the expression of Nrf2 and HO-1, which are cellular antioxidant proteins. Reactive Oxygen Species 45-68 heme oxygenase 1 Homo sapiens 119-123 30367621-4 2018 Since the degradation of Bach1 protein relies on protein phosphorylation, we hypothesized that DHA-induced HO-1 gene transcription could be attenuated by kinase inhibitors, resulting in an enhanced cytotoxicity. Docosahexaenoic Acids 95-98 heme oxygenase 1 Homo sapiens 107-111 30367621-11 2018 Consequently, DHA-induced HO-1 gene transcription was reversed by Sorafenib as evidenced by western blot and reporter gene analysis. Docosahexaenoic Acids 14-17 heme oxygenase 1 Homo sapiens 26-30 30367621-11 2018 Consequently, DHA-induced HO-1 gene transcription was reversed by Sorafenib as evidenced by western blot and reporter gene analysis. Sorafenib 66-75 heme oxygenase 1 Homo sapiens 26-30 30367621-14 2018 CONCLUSIONS: We demonstrate that sorafenib attenuates DHA-induced HO-1 expression and acts in synergy with DHA to suppress cancer cell viability and tumor growth. Sorafenib 33-42 heme oxygenase 1 Homo sapiens 66-70 30367621-14 2018 CONCLUSIONS: We demonstrate that sorafenib attenuates DHA-induced HO-1 expression and acts in synergy with DHA to suppress cancer cell viability and tumor growth. Docosahexaenoic Acids 54-57 heme oxygenase 1 Homo sapiens 66-70 30416532-5 2018 Additionally, exposure of bone marrow stromal cells (HS-5) to NAC increased adiponectin, PPARgamma, HO-1, and SIRT-1 and increased beta-oxidation markers such as PPARalpha and PPARdelta mRNA levels. Acetylcysteine 62-65 heme oxygenase 1 Homo sapiens 100-104 30397576-3 2018 The potency of activation is dependent on the leaving group at the 3-position of the isoxazoline nucleus, and an additional ring on the molecule limits the Nrf2/HO-1 activating properties. Isoxazoline 85-96 heme oxygenase 1 Homo sapiens 161-165 30144404-7 2018 Glutamine deprivation or GLS1 inhibition also stimulated the production of reactive oxygen species and this was associated with a marked decline in heme oxygenase-1 (HO-1) expression. Glutamine 0-9 heme oxygenase 1 Homo sapiens 148-164 30105448-13 2018 The induction of HO-1 might have be a promising approach for the treatment of psoriasis through antioxidant ability, immunomodulatory role as well as its role in heme synthesis. Heme 162-166 heme oxygenase 1 Homo sapiens 17-21 30261468-0 2018 Targeting heme Oxygenase-1 with hybrid compounds to overcome Imatinib resistance in chronic myeloid leukemia cell lines. Imatinib Mesylate 61-69 heme oxygenase 1 Homo sapiens 10-26 30261468-4 2018 With this in mind, in this study, we designed and synthesized the first series of hybrid compounds obtained by combining the structures of IM, as BCR-ABL inhibitor, with imidazole-based HO-1 inhibitors. imidazole 170-179 heme oxygenase 1 Homo sapiens 186-190 30349652-2 2018 The cytoprotective enzyme heme oxygenase-1 (HO-1) has been shown to mediate anti-proliferative and anti-migratory effects in VSMC. vsmc 125-129 heme oxygenase 1 Homo sapiens 26-42 30349652-2 2018 The cytoprotective enzyme heme oxygenase-1 (HO-1) has been shown to mediate anti-proliferative and anti-migratory effects in VSMC. vsmc 125-129 heme oxygenase 1 Homo sapiens 44-48 30349652-3 2018 This study investigates the effect of cannabidiol (CBD), a non-psychoactive cannabinoid, on HO-1 expression and disease-associated functions of human umbilical artery smooth muscle cells (HUASMC). Cannabidiol 38-49 heme oxygenase 1 Homo sapiens 92-96 30349652-3 2018 This study investigates the effect of cannabidiol (CBD), a non-psychoactive cannabinoid, on HO-1 expression and disease-associated functions of human umbilical artery smooth muscle cells (HUASMC). Cannabidiol 51-54 heme oxygenase 1 Homo sapiens 92-96 30349652-6 2018 Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. Glutathione 71-82 heme oxygenase 1 Homo sapiens 38-42 30349652-6 2018 Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. Acetylcysteine 93-109 heme oxygenase 1 Homo sapiens 38-42 30349652-6 2018 Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. Reactive Oxygen Species 143-166 heme oxygenase 1 Homo sapiens 38-42 30349652-6 2018 Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. Reactive Oxygen Species 168-171 heme oxygenase 1 Homo sapiens 38-42 30349652-6 2018 Instead, the CBD-mediated increase in HO-1 protein was reversed by the glutathione precursor N-acetylcysteine, indicating the participation of reactive oxygen species (ROS) signaling; this was confirmed by flow cytometry-based ROS detection. Reactive Oxygen Species 227-230 heme oxygenase 1 Homo sapiens 38-42 30349652-10 2018 Collectively, this work provides the first indication of CBD-mediated enhancement of HO-1 in VSMC and potential protective effects against aberrant VSMC proliferation and migration. Cannabidiol 57-60 heme oxygenase 1 Homo sapiens 85-89 30144404-7 2018 Glutamine deprivation or GLS1 inhibition also stimulated the production of reactive oxygen species and this was associated with a marked decline in heme oxygenase-1 (HO-1) expression. Glutamine 0-9 heme oxygenase 1 Homo sapiens 166-170 30144404-8 2018 GLS1 inhibition also sensitized ECs to the cytotoxic effect of hydrogen peroxide and this was prevented by the overexpression of HO-1. Hydrogen Peroxide 63-80 heme oxygenase 1 Homo sapiens 129-133 30088830-1 2018 BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) is induced by thiazolidinediones including rosiglitazone and exerts anti-inflammatory effects in various models. Thiazolidinediones 62-80 heme oxygenase 1 Homo sapiens 24-40 30088830-0 2018 Haem oxygenase-1 up-regulation by rosiglitazone via ROS-dependent Nrf2-antioxidant response elements axis or PPARgamma attenuates LPS-mediated lung inflammation. Rosiglitazone 34-47 heme oxygenase 1 Homo sapiens 0-16 30088830-1 2018 BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) is induced by thiazolidinediones including rosiglitazone and exerts anti-inflammatory effects in various models. Thiazolidinediones 62-80 heme oxygenase 1 Homo sapiens 42-46 30088830-0 2018 Haem oxygenase-1 up-regulation by rosiglitazone via ROS-dependent Nrf2-antioxidant response elements axis or PPARgamma attenuates LPS-mediated lung inflammation. Rosiglitazone 52-55 heme oxygenase 1 Homo sapiens 0-16 30088830-1 2018 BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) is induced by thiazolidinediones including rosiglitazone and exerts anti-inflammatory effects in various models. Rosiglitazone 91-104 heme oxygenase 1 Homo sapiens 24-40 30088830-1 2018 BACKGROUND AND PURPOSE: Haem oxygenase-1 (HO-1) is induced by thiazolidinediones including rosiglitazone and exerts anti-inflammatory effects in various models. Rosiglitazone 91-104 heme oxygenase 1 Homo sapiens 42-46 30088830-2 2018 However, the molecular mechanisms underlying rosiglitazone-induced HO-1 expression remain largely unknown in human pulmonary alveolar epithelial cells (HPAEpiCs). Rosiglitazone 45-58 heme oxygenase 1 Homo sapiens 67-71 30085341-0 2018 Cyclooxygenase-2-mediated upregulation of heme oxygenase 1 mitigates the toxicity of deuterium-tritium fusion radiation. deuterium-tritium 85-102 heme oxygenase 1 Homo sapiens 42-58 29748730-9 2018 The expression of Nrf2 and HO-1 was up-regulated by the treatment of corynoline. corynoline 69-79 heme oxygenase 1 Homo sapiens 27-31 30085341-6 2018 Pretreatment with the HO-1 inhibitor, protoporphyrin IX zinc (II), exacerbated double strand break formation following exposure to fusion radiation. protoporphyrin IX 38-55 heme oxygenase 1 Homo sapiens 22-26 30085341-7 2018 The expression of cyclooxygenase-2 (COX-2) contributed to the upregulation of HO-1, as demonstrated by the result that its inhibitor, NS-398, inhibited the induction of HO-1 in irradiated cells. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 134-140 heme oxygenase 1 Homo sapiens 78-82 30085341-7 2018 The expression of cyclooxygenase-2 (COX-2) contributed to the upregulation of HO-1, as demonstrated by the result that its inhibitor, NS-398, inhibited the induction of HO-1 in irradiated cells. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 134-140 heme oxygenase 1 Homo sapiens 169-173 29959986-6 2018 Long-term iron exposure resulted in iron accumulation, cytosolic ROS formation and increased heme oxygenase 1 (HMOX-1) mRNA expression (all p < 0.001). Iron 10-14 heme oxygenase 1 Homo sapiens 93-109 29797346-0 2018 Hemin provides protection against lead neurotoxicity through heme oxygenase 1/carbon monoxide activation. Hemin 0-5 heme oxygenase 1 Homo sapiens 61-77 29797346-3 2018 In this study, we applied hemin, the substrate and a well-known inducer of HO-1, to investigate the possible role of HO-1 in protecting against Pb neurotoxicity. Hemin 26-31 heme oxygenase 1 Homo sapiens 75-79 29797346-3 2018 In this study, we applied hemin, the substrate and a well-known inducer of HO-1, to investigate the possible role of HO-1 in protecting against Pb neurotoxicity. Lead 144-146 heme oxygenase 1 Homo sapiens 117-121 29797346-6 2018 However, knocking down HO-1 could significantly abolish the cytoprotective action of hemin against Pb toxicity, confirming HO-1 contributed to the protection. Hemin 85-90 heme oxygenase 1 Homo sapiens 23-27 29797346-6 2018 However, knocking down HO-1 could significantly abolish the cytoprotective action of hemin against Pb toxicity, confirming HO-1 contributed to the protection. Hemin 85-90 heme oxygenase 1 Homo sapiens 123-127 29797346-6 2018 However, knocking down HO-1 could significantly abolish the cytoprotective action of hemin against Pb toxicity, confirming HO-1 contributed to the protection. Lead 99-101 heme oxygenase 1 Homo sapiens 23-27 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 13-17 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 123-127 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Hemin 150-155 heme oxygenase 1 Homo sapiens 13-17 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Hemin 150-155 heme oxygenase 1 Homo sapiens 123-127 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Lead 174-176 heme oxygenase 1 Homo sapiens 13-17 29797346-7 2018 Finally, the HO-1-derived production of carbon monoxide, but not of bilirubin or Fe2+ , mediated the protective effects of HO-1 activation induced by hemin treatment against Pb-induced cell death and oxidative stress in SHSY5Y cells. Lead 174-176 heme oxygenase 1 Homo sapiens 123-127 29797346-8 2018 Overall, this study showed that hemin provided protection against Pb neurotoxicity by HO-1/carbon monoxide activation. Hemin 32-37 heme oxygenase 1 Homo sapiens 86-90 29797346-8 2018 Overall, this study showed that hemin provided protection against Pb neurotoxicity by HO-1/carbon monoxide activation. Lead 66-68 heme oxygenase 1 Homo sapiens 86-90 30216854-7 2018 We found that RS9 protected ARPE-19 cells against NaIO3-induced oxidative damage, and that the protective effects of RS9 were inhibited by co-treatment with zinc protoporphyrin, an HO-1 inhibitor. zinc protoporphyrin 157-176 heme oxygenase 1 Homo sapiens 181-185 29882686-10 2018 ZnO and CuO nanoparticles displayed severe cytotoxicity and enhanced gene expression of heme oxygenase-1 (HO-1) and interleukin-8 (IL-8). Zinc Oxide 0-3 heme oxygenase 1 Homo sapiens 88-104 29882686-10 2018 ZnO and CuO nanoparticles displayed severe cytotoxicity and enhanced gene expression of heme oxygenase-1 (HO-1) and interleukin-8 (IL-8). Zinc Oxide 0-3 heme oxygenase 1 Homo sapiens 106-110 29882686-14 2018 The enhancement of HO-1, IL-8, and MT2A gene expressions was related to the cytotoxic activity of metal oxide nanoparticles. metal oxide 98-109 heme oxygenase 1 Homo sapiens 19-23 30040983-0 2018 Heme Oxygenase-1 protects astroglia against manganese-induced oxidative injury by regulating mitochondrial quality control. Manganese 44-53 heme oxygenase 1 Homo sapiens 0-16 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Heme 69-73 heme oxygenase 1 Homo sapiens 0-16 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Heme 69-73 heme oxygenase 1 Homo sapiens 18-22 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Iron 91-95 heme oxygenase 1 Homo sapiens 0-16 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Iron 91-95 heme oxygenase 1 Homo sapiens 18-22 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 0-16 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 18-22 30214615-7 2018 The present study demonstrated a positive correlation between 8-OHdG and HO-1 levels (P=0.012). 8-ohdg 62-68 heme oxygenase 1 Homo sapiens 73-77 29530745-1 2018 OBJECTIVE: The aim of this study was to investigate how maternal cell-free fetal hemoglobin and heme impact the scavenger enzyme systems Hemopexin and Heme Oxygenase-1 in patients with preeclampsia (PE). Heme 96-100 heme oxygenase 1 Homo sapiens 151-167 29530745-10 2018 CONCLUSIONS: Increased maternal plasma levels of heme and HbF in PE are associated with decreased HO-1 and hemopexin protein levels as well as reduced hemopexin activity. Heme 49-53 heme oxygenase 1 Homo sapiens 98-102 30031050-7 2018 In Drp1 knock out cells, HO-1 levels were low compared to wild type cells, both in untreated controls as well as after BH3I-1 exposure, demonstrating that Drp1 is at least in part required for determining basal and inducible HO-1 levels. BH3I-1 119-125 heme oxygenase 1 Homo sapiens 25-29 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Biliverdine 118-128 heme oxygenase 1 Homo sapiens 0-16 29959986-6 2018 Long-term iron exposure resulted in iron accumulation, cytosolic ROS formation and increased heme oxygenase 1 (HMOX-1) mRNA expression (all p < 0.001). Iron 10-14 heme oxygenase 1 Homo sapiens 111-117 30040983-1 2018 Heme Oxygenase-1 (HO-1), a stress- responsive enzyme which catalyzes heme degradation into iron, carbon monoxide, and biliverdin, exerts a neuroprotective role involving many different signaling pathways. Biliverdine 118-128 heme oxygenase 1 Homo sapiens 18-22 30327711-12 2018 Treatment of cultured human artery endothelial cells with curcumin induced the HO-1 expression through the activation of nuclear factor-E2-related factor 2 (Nrf2) and an antioxidant responsive element via the p38 MAPK signalling pathway. Curcumin 58-66 heme oxygenase 1 Homo sapiens 79-83 30040983-4 2018 HO-1 exerted a protective effect against Mn2+ injury. Manganese(2+) 41-45 heme oxygenase 1 Homo sapiens 0-4 30040983-5 2018 In fact, HO-1 decreased both intracellular and mitochondrial reactive oxygen species as well as the appearance of apoptotic features. Reactive Oxygen Species 61-84 heme oxygenase 1 Homo sapiens 9-13 30040983-6 2018 Considering that Mn2+ induces mitochondrial damage and a defective MQC has been implicated in neurodegenerative diseases, we hypothesized that HO-1 could mediate cytoprotection by regulating the MQC processes. Manganese(2+) 17-21 heme oxygenase 1 Homo sapiens 143-147 30040983-8 2018 Altogether, our data demonstrate a pro-survival function for HO-1 in Mn2+-induced apoptosis that involves the preservation of a proper MQC. Manganese(2+) 69-73 heme oxygenase 1 Homo sapiens 61-65 30055129-8 2018 All of our data confirmed that oxidative stress-mediated mitochondrial apoptosis (especially the Nrf-2/HO-1 pathway) is responsible for 5h2c-induced HCC damage. 5h2c 136-140 heme oxygenase 1 Homo sapiens 103-107 30956868-4 2019 The treatment with chebulic acid attenuated cell death in t-BHP-induced HepG2 liver cells and increased intracellular glutathione content, upregulated the activity of heme oxygenase-1, and also increased the translocation of Nrf2 into the nucleus and Nrf2 target gene expression in a dose-dependent manner. chebulic acid 19-32 heme oxygenase 1 Homo sapiens 167-183 30053409-0 2018 Hydroxytyrosol butyrate inhibits 6-OHDA-induced apoptosis through activation of the Nrf2/HO-1 axis in SH-SY5Y cells. 2-(3,4-Dihydroxyphenyl)ethyl butyrate 0-23 heme oxygenase 1 Homo sapiens 89-93 30218018-6 2018 We validated genes belonging to these pathways, such as HSPA5, SEC61B, G6PD, HMOX1 and PDE3B to be cooperatively modulated by the OPCs-curcumin combination. Curcumin 135-143 heme oxygenase 1 Homo sapiens 77-82 30327713-0 2018 Heme Oxygenase-1 May Affect Cell Signalling via Modulation of Ganglioside Composition. Gangliosides 62-73 heme oxygenase 1 Homo sapiens 0-16 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Heme 56-60 heme oxygenase 1 Homo sapiens 0-16 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Heme 56-60 heme oxygenase 1 Homo sapiens 18-23 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 0-16 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 18-23 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Iron 81-85 heme oxygenase 1 Homo sapiens 0-16 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Iron 81-85 heme oxygenase 1 Homo sapiens 18-23 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 0-16 30327713-1 2018 Heme oxygenase 1 (Hmox1), a ubiquitous enzyme degrading heme to carbon monoxide, iron, and biliverdin, is one of the cytoprotective enzymes induced in response to a variety of stimuli, including cellular oxidative stress. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 18-23 30327713-4 2018 The aim of this study was to assess the possible role of Hmox1 in ganglioside metabolism in relation to oxidative stress. Gangliosides 66-77 heme oxygenase 1 Homo sapiens 57-62 30327713-6 2018 To elucidate the possible underlying mechanisms between Hmox1 and ganglioside metabolism, hepatoblastoma HepG2 and neuroblastoma SH-SY5Y cell lines were used for in vitro experiments. Gangliosides 66-77 heme oxygenase 1 Homo sapiens 56-61 30258436-1 2018 Heme Oxygenase 1 (HMOX1) is an enzyme that catalyzes the reaction that degrades the heme group contained in several important proteins, such as hemoglobin, myoglobin, and cytochrome p450. Heme 84-88 heme oxygenase 1 Homo sapiens 0-16 30258436-1 2018 Heme Oxygenase 1 (HMOX1) is an enzyme that catalyzes the reaction that degrades the heme group contained in several important proteins, such as hemoglobin, myoglobin, and cytochrome p450. Heme 84-88 heme oxygenase 1 Homo sapiens 18-23 30258436-2 2018 The enzymatic reaction catalyzed by HMOX1 generates Fe2+, biliverdin and CO. ammonium ferrous sulfate 52-56 heme oxygenase 1 Homo sapiens 36-41 30258436-2 2018 The enzymatic reaction catalyzed by HMOX1 generates Fe2+, biliverdin and CO. Biliverdine 58-68 heme oxygenase 1 Homo sapiens 36-41 30258436-2 2018 The enzymatic reaction catalyzed by HMOX1 generates Fe2+, biliverdin and CO. Carbon Monoxide 73-75 heme oxygenase 1 Homo sapiens 36-41 30053409-0 2018 Hydroxytyrosol butyrate inhibits 6-OHDA-induced apoptosis through activation of the Nrf2/HO-1 axis in SH-SY5Y cells. Oxidopamine 33-39 heme oxygenase 1 Homo sapiens 89-93 30053409-4 2018 HT-B also induced the protein expression of the transcription factor, NF-E2-related factor-2 (Nrf2) and its transcriptional activation, resulting in the up-regulated expression of heme oxygenase-1 (HO-1), which conferred neuroprotection against 6-OHDA-induced oxidative damage. Oxidopamine 245-251 heme oxygenase 1 Homo sapiens 180-196 30053409-4 2018 HT-B also induced the protein expression of the transcription factor, NF-E2-related factor-2 (Nrf2) and its transcriptional activation, resulting in the up-regulated expression of heme oxygenase-1 (HO-1), which conferred neuroprotection against 6-OHDA-induced oxidative damage. Oxidopamine 245-251 heme oxygenase 1 Homo sapiens 198-202 29761622-4 2018 HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. Heme 47-51 heme oxygenase 1 Homo sapiens 0-5 29671417-5 2018 Furthermore, pretreatment with cyanidin greatly promoted the translocation of the Nrf2 protein from the cytoplasm to the nucleus; upregulating cytoprotective enzymes, including HO-1, NQO-1 and GCLC; and increased the activity of SOD enzymes. cyanidin 31-39 heme oxygenase 1 Homo sapiens 177-181 29913410-10 2018 Notably, IA increased the antioxidant enzyme heme oxygenase-1 (HO-1) expression and this effect was reversed by HO-1 inhibitor zinc protoporphyrin (ZnPP) leading to reduction of the neuroprotective effect of IA against Abeta-induced neurotoxicity. zinc protoporphyrin 127-146 heme oxygenase 1 Homo sapiens 45-61 29913410-10 2018 Notably, IA increased the antioxidant enzyme heme oxygenase-1 (HO-1) expression and this effect was reversed by HO-1 inhibitor zinc protoporphyrin (ZnPP) leading to reduction of the neuroprotective effect of IA against Abeta-induced neurotoxicity. zinc protoporphyrin 148-152 heme oxygenase 1 Homo sapiens 45-61 29947925-3 2018 Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Glutathione 284-295 heme oxygenase 1 Homo sapiens 179-195 29947925-3 2018 Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Glutathione 284-295 heme oxygenase 1 Homo sapiens 197-201 29947925-3 2018 Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Glutathione 297-300 heme oxygenase 1 Homo sapiens 179-195 29947925-3 2018 Nuclear factor erythroid-derived 2-like 2 (NFE2L2 or Nrf-2) is essential for the antioxidant responsive element (ARE)-mediated induction of endogenous antioxidant enzymes such as heme oxygenase 1 (HO-1) and glutamate-cysteine ligase [GCL, the rate-limiting enzyme in the synthesis of glutathione (GSH)]. Glutathione 297-300 heme oxygenase 1 Homo sapiens 197-201 29066411-5 2018 Additionally, GA induced phosphorylated activation of extracellular regulated kinase (ERK), and ERK inhibitor PD98059 partially decreased GA-induced hepatoprotection, and downregulated the increased protein expressions of Nrf2, GCLC and HO-1 induced by GA. Gallic Acid 14-16 heme oxygenase 1 Homo sapiens 237-241 29066411-5 2018 Additionally, GA induced phosphorylated activation of extracellular regulated kinase (ERK), and ERK inhibitor PD98059 partially decreased GA-induced hepatoprotection, and downregulated the increased protein expressions of Nrf2, GCLC and HO-1 induced by GA. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 110-117 heme oxygenase 1 Homo sapiens 237-241 30186615-9 2018 Results: Pretreatment with low-dose (1 and 5 muM) CAM for 72 h inhibited H2O2-induced reductions of GPx-1, GR, SOD, CAT and HO-1 activities, and mRNA expressions of GPx-1 and HO-1, and improved the GSH/GSSG ratio. Hydrogen Peroxide 73-77 heme oxygenase 1 Homo sapiens 124-128 30186615-9 2018 Results: Pretreatment with low-dose (1 and 5 muM) CAM for 72 h inhibited H2O2-induced reductions of GPx-1, GR, SOD, CAT and HO-1 activities, and mRNA expressions of GPx-1 and HO-1, and improved the GSH/GSSG ratio. Hydrogen Peroxide 73-77 heme oxygenase 1 Homo sapiens 175-179 29761622-4 2018 HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 0-5 29761622-4 2018 HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. Carbon Monoxide 81-83 heme oxygenase 1 Homo sapiens 0-5 29761622-4 2018 HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. Biliverdine 86-96 heme oxygenase 1 Homo sapiens 0-5 29761622-4 2018 HMOX1 is a cytoprotective enzyme that degrades heme to generate carbon monoxide (CO), biliverdin, and molecular iron. Iron 112-116 heme oxygenase 1 Homo sapiens 0-5 29974998-6 2018 Moreover, the expression of HO-1, a functional modulator of Tregs, was decreased in vitiligo Tregs, and the concentrations of HO-1 metabolites, including bilirubin, CoHb and iron, were correspondingly decreased in serum of vitiligo patients. Bilirubin 154-163 heme oxygenase 1 Homo sapiens 126-130 29974998-6 2018 Moreover, the expression of HO-1, a functional modulator of Tregs, was decreased in vitiligo Tregs, and the concentrations of HO-1 metabolites, including bilirubin, CoHb and iron, were correspondingly decreased in serum of vitiligo patients. Iron 174-178 heme oxygenase 1 Homo sapiens 28-32 30015902-7 2018 Furthermore, spilanthol significantly suppressed the phosphorylation of c-Jun N-terminal kinase (JNK), while pretreatment with spilanthol enhanced heme oxygenase (HO)-1 protein expression by western blotting. N-isobutyl-2E-decenamide 127-137 heme oxygenase 1 Homo sapiens 147-168 29278009-1 2018 BACKGROUND: Heme oxygenase (HO)-1 is a heme-degrading enzyme highly expressed in monocyte/macrophage, serum levels of which may be promising biomarker for adult-onset Still"s disease (AOSD). Heme 39-43 heme oxygenase 1 Homo sapiens 12-33 30302047-7 2018 More recent studies also suggest a role of ROS-induced downregulation of Heme Oxygenase (HO)1-targeting micro RNAs and upregulation of HO1 for ammonia-induced proliferation inhibition in cultured astrocytes. Ammonia 143-150 heme oxygenase 1 Homo sapiens 135-138 30015902-0 2018 Spilanthol inhibits TNF-alpha-induced ICAM-1 expression and pro-inflammatory responses by inducing heme oxygenase-1 expression and suppressing pJNK in HaCaT keratinocytes. N-isobutyl-2E-decenamide 0-10 heme oxygenase 1 Homo sapiens 99-115 30122195-4 2018 l-Arg deprivation markedly increased the mRNA expression of heat shock protein 70 and heme-oxygenase-1 under HS conditions. Arginine 0-5 heme oxygenase 1 Homo sapiens 86-102 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Heme 75-79 heme oxygenase 1 Homo sapiens 24-40 30041041-10 2018 Notably, activation of HO-1 gene expression further increased the levels of EETs, suggesting that the antioxidant HO-1 system protects EETs from degradation by ROS. ros 160-163 heme oxygenase 1 Homo sapiens 23-27 30041041-10 2018 Notably, activation of HO-1 gene expression further increased the levels of EETs, suggesting that the antioxidant HO-1 system protects EETs from degradation by ROS. ros 160-163 heme oxygenase 1 Homo sapiens 114-118 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Heme 75-79 heme oxygenase 1 Homo sapiens 42-46 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 24-40 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 42-46 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 24-40 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 42-46 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Iron 125-129 heme oxygenase 1 Homo sapiens 24-40 30354985-1 2018 Background and Purpose- Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Iron 125-129 heme oxygenase 1 Homo sapiens 42-46 30159112-9 2018 Trx, nuclear Nrf2 protein, and HO-1 protein were also negatively correlated with the percent of time that SaO2 was less than 90% (TSat90). sao2 106-110 heme oxygenase 1 Homo sapiens 31-35 29969714-5 2018 We find that, in combination with 2.5 microM sulforaphane, dtBHQ markedly enhances ARE-regulated gene expression, including expression of the cytoprotective proteins aldo-keto reductase family 1 member C1 (AKR1C1) and heme oxygenase-1 (HO-1). 2,5-di-tert-butylhydroquinone 59-64 heme oxygenase 1 Homo sapiens 218-234 29753693-7 2018 The combination of ALA 600 mg and sodium ferrous citrate (SFC) 942 mg upregulated HO-1 in PBMC at 8 h after administration while sole administration of ALA or SFC was unable to induce HO-1. 5-amino levulinic acid 19-22 heme oxygenase 1 Homo sapiens 82-86 29753693-7 2018 The combination of ALA 600 mg and sodium ferrous citrate (SFC) 942 mg upregulated HO-1 in PBMC at 8 h after administration while sole administration of ALA or SFC was unable to induce HO-1. Ferromia 34-56 heme oxygenase 1 Homo sapiens 82-86 29753693-7 2018 The combination of ALA 600 mg and sodium ferrous citrate (SFC) 942 mg upregulated HO-1 in PBMC at 8 h after administration while sole administration of ALA or SFC was unable to induce HO-1. SFC 58-61 heme oxygenase 1 Homo sapiens 82-86 29753693-8 2018 HO-1 in blood myeloid and plasmacytoid dendritic cells was also upregulated with ALA+SFC. ala+sfc 81-88 heme oxygenase 1 Homo sapiens 0-4 29993075-0 2018 Targeting heme oxygenase-1 by quercetin ameliorates alcohol-induced acute liver injury via inhibiting NLRP3 inflammasome activation. Quercetin 30-39 heme oxygenase 1 Homo sapiens 10-26 29993075-0 2018 Targeting heme oxygenase-1 by quercetin ameliorates alcohol-induced acute liver injury via inhibiting NLRP3 inflammasome activation. Alcohols 52-59 heme oxygenase 1 Homo sapiens 10-26 29993075-7 2018 Moreover, this protective effect of quercetin could be diminished when combined with the HO-1 inhibitor ZnppIX which demonstrated a critical role of HO-1 in quercetin"s hepatoprotection. Quercetin 36-45 heme oxygenase 1 Homo sapiens 149-153 29993075-7 2018 Moreover, this protective effect of quercetin could be diminished when combined with the HO-1 inhibitor ZnppIX which demonstrated a critical role of HO-1 in quercetin"s hepatoprotection. zinc protoporphyrin 104-110 heme oxygenase 1 Homo sapiens 89-93 29993075-7 2018 Moreover, this protective effect of quercetin could be diminished when combined with the HO-1 inhibitor ZnppIX which demonstrated a critical role of HO-1 in quercetin"s hepatoprotection. zinc protoporphyrin 104-110 heme oxygenase 1 Homo sapiens 149-153 29993075-7 2018 Moreover, this protective effect of quercetin could be diminished when combined with the HO-1 inhibitor ZnppIX which demonstrated a critical role of HO-1 in quercetin"s hepatoprotection. Quercetin 157-166 heme oxygenase 1 Homo sapiens 89-93 29993075-7 2018 Moreover, this protective effect of quercetin could be diminished when combined with the HO-1 inhibitor ZnppIX which demonstrated a critical role of HO-1 in quercetin"s hepatoprotection. Quercetin 157-166 heme oxygenase 1 Homo sapiens 149-153 29993075-8 2018 The underlying mechanism of quercetin"s benefit on the liver may be explained by its anti-oxidant properties and inhibitory effect on the ROS/NF-kappaB/NLRP3 inflammasome/IL-1beta and IL-18 pathway by inducing HO-1. Quercetin 28-37 heme oxygenase 1 Homo sapiens 210-214 29993075-10 2018 In conclusion, quercetin can preserve the function of the liver in acute alcoholic injury by upregulating the expression of IL-10 and HO-1 and thus inhibiting NLRP3 inflammasome activation and inflammatory factor secretion. Quercetin 15-24 heme oxygenase 1 Homo sapiens 134-138 29777873-4 2018 Moreover, cinnamaldehyde inhibited the phosphorylation of p38, JNK and p65 NF-kappaB and increased heme oxygenase-1 (HO-1) activity. cinnamaldehyde 10-24 heme oxygenase 1 Homo sapiens 99-115 29777873-4 2018 Moreover, cinnamaldehyde inhibited the phosphorylation of p38, JNK and p65 NF-kappaB and increased heme oxygenase-1 (HO-1) activity. cinnamaldehyde 10-24 heme oxygenase 1 Homo sapiens 117-121 29777873-7 2018 Thus, results indicated that cinnamaldehyde antagonized the ox-LDL-induced VSMCs proliferation, migration, inflammation and foam cell formation through regulation of HO-1, MMP-2, LOX-1 and blockage of cell cycle, and - suppression of p38, JNK/MAPK and NF-kappaB signaling pathways. cinnamaldehyde 29-43 heme oxygenase 1 Homo sapiens 166-170 30153269-7 2018 These data suggest that the HO-1-derived metabolites, CO and bilirubin, elevate astrocytic mitochondrial function via a HIF-1alpha/ERRalpha circuit coupled with L-type Ca2+ channel activation and PGC-1alpha-mediated oxygen consumption. Carbon Monoxide 54-56 heme oxygenase 1 Homo sapiens 28-32 30153269-7 2018 These data suggest that the HO-1-derived metabolites, CO and bilirubin, elevate astrocytic mitochondrial function via a HIF-1alpha/ERRalpha circuit coupled with L-type Ca2+ channel activation and PGC-1alpha-mediated oxygen consumption. Bilirubin 61-70 heme oxygenase 1 Homo sapiens 28-32 30153269-7 2018 These data suggest that the HO-1-derived metabolites, CO and bilirubin, elevate astrocytic mitochondrial function via a HIF-1alpha/ERRalpha circuit coupled with L-type Ca2+ channel activation and PGC-1alpha-mediated oxygen consumption. Oxygen 216-222 heme oxygenase 1 Homo sapiens 28-32 30061089-4 2018 In this study, we found that increased expression of Mkp-1, Nrf2, and heme oxygenase 1 (Ho-1) was correlated in colonic tissues in patients with ulcerative colitis and Crohn"s disease, as well as wild-type mice with colitis induced by dextran sodium sulfate (DSS). dextran sodium sulfate 235-257 heme oxygenase 1 Homo sapiens 70-86 30061089-4 2018 In this study, we found that increased expression of Mkp-1, Nrf2, and heme oxygenase 1 (Ho-1) was correlated in colonic tissues in patients with ulcerative colitis and Crohn"s disease, as well as wild-type mice with colitis induced by dextran sodium sulfate (DSS). dextran sodium sulfate 235-257 heme oxygenase 1 Homo sapiens 88-92 30061089-4 2018 In this study, we found that increased expression of Mkp-1, Nrf2, and heme oxygenase 1 (Ho-1) was correlated in colonic tissues in patients with ulcerative colitis and Crohn"s disease, as well as wild-type mice with colitis induced by dextran sodium sulfate (DSS). dss 259-262 heme oxygenase 1 Homo sapiens 70-86 30061089-4 2018 In this study, we found that increased expression of Mkp-1, Nrf2, and heme oxygenase 1 (Ho-1) was correlated in colonic tissues in patients with ulcerative colitis and Crohn"s disease, as well as wild-type mice with colitis induced by dextran sodium sulfate (DSS). dss 259-262 heme oxygenase 1 Homo sapiens 88-92 29753693-0 2018 Oral administration of 5-aminolevulinic acid induces heme oxygenase-1 expression in peripheral blood mononuclear cells of healthy human subjects in combination with ferrous iron. 5-amino levulinic acid 23-44 heme oxygenase 1 Homo sapiens 53-69 29753693-2 2018 The aim of this study is to determine whether the combination of 5-aminolevulinic acid (ALA) and iron induces HO-1 expression in healthy human peripheral blood mononuclear cells (PBMC). 5-amino levulinic acid 65-86 heme oxygenase 1 Homo sapiens 110-114 29753693-2 2018 The aim of this study is to determine whether the combination of 5-aminolevulinic acid (ALA) and iron induces HO-1 expression in healthy human peripheral blood mononuclear cells (PBMC). 5-amino levulinic acid 88-91 heme oxygenase 1 Homo sapiens 110-114 29753693-2 2018 The aim of this study is to determine whether the combination of 5-aminolevulinic acid (ALA) and iron induces HO-1 expression in healthy human peripheral blood mononuclear cells (PBMC). Iron 97-101 heme oxygenase 1 Homo sapiens 110-114 29753693-5 2018 Study B aimed to examine HO-1 dose dependency at 150, 300, and 600 mg of ALA and the need for iron supplementation. 5-amino levulinic acid 73-76 heme oxygenase 1 Homo sapiens 25-29 29753693-6 2018 Study C aimed to investigate HO-1 changes during a three-day, repetitive administration of ALA and iron. 5-amino levulinic acid 91-94 heme oxygenase 1 Homo sapiens 29-33 29753693-6 2018 Study C aimed to investigate HO-1 changes during a three-day, repetitive administration of ALA and iron. Iron 99-103 heme oxygenase 1 Homo sapiens 29-33 30092096-11 2018 The mRNA levels of heme oxygenase-1 and superoxide dismutase-1 and ROS levels were significantly increased in HGFs after 72 h of exposure to 50 mM glucose concentration. Glucose 147-154 heme oxygenase 1 Homo sapiens 19-35 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Heme 89-93 heme oxygenase 1 Homo sapiens 6-22 30096430-0 2018 WITHDRAWN: Paeoniflorin attenuates LPS-induced inflammation in nucleus pulposus cells via Nrf-2/HO-1/HMGB1/NF-kappaB pathway. peoniflorin 11-23 heme oxygenase 1 Homo sapiens 96-100 29961319-6 2018 Further investigations showed that PSB significantly enhanced Nrf2 expression and nuclear accumulation, improved ARE promoter activity and up-regulated expression of HO-1 and GCLC. pinostrobin 35-38 heme oxygenase 1 Homo sapiens 166-170 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Heme 89-93 heme oxygenase 1 Homo sapiens 24-28 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Carbon Monoxide 106-108 heme oxygenase 1 Homo sapiens 6-22 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Carbon Monoxide 106-108 heme oxygenase 1 Homo sapiens 24-28 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Biliverdine 110-120 heme oxygenase 1 Homo sapiens 6-22 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Biliverdine 110-120 heme oxygenase 1 Homo sapiens 24-28 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Iron 126-130 heme oxygenase 1 Homo sapiens 6-22 30159103-1 2018 Aims: Heme oxygenase-1 (HO-1) is an intracellular enzyme that catalyzes the oxidation of heme to generate CO, biliverdin, and iron. Iron 126-130 heme oxygenase 1 Homo sapiens 24-28 30186543-4 2018 We found that the antioxidant cinnamaldehyde activated the NRF2/HMOX1 pathway. cinnamaldehyde 30-44 heme oxygenase 1 Homo sapiens 64-69 29969038-6 2018 Moreover, metformin alleviated LPS-induced NF-kappaB phosphorylation, promoted Nrf2 nuclear translocation, and increased the expression of the antioxidative genes (HO-1 and NQO-1), leading to reduced intestinal ROS content. Metformin 10-19 heme oxygenase 1 Homo sapiens 164-168 29730426-6 2018 Mechanism studies demonstrated that isorhamnetin pretreatment remarkably abolished H/R-induced downregulation of Nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions and upregulation of NADPH oxidase-2/4 (NOX-2/4) expressions in cardiomyocytes. r 85-86 heme oxygenase 1 Homo sapiens 168-184 29730426-6 2018 Mechanism studies demonstrated that isorhamnetin pretreatment remarkably abolished H/R-induced downregulation of Nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions and upregulation of NADPH oxidase-2/4 (NOX-2/4) expressions in cardiomyocytes. r 85-86 heme oxygenase 1 Homo sapiens 186-190 29730426-6 2018 Mechanism studies demonstrated that isorhamnetin pretreatment remarkably abolished H/R-induced downregulation of Nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions and upregulation of NADPH oxidase-2/4 (NOX-2/4) expressions in cardiomyocytes. 3-methylquercetin 36-48 heme oxygenase 1 Homo sapiens 168-184 29730426-6 2018 Mechanism studies demonstrated that isorhamnetin pretreatment remarkably abolished H/R-induced downregulation of Nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions and upregulation of NADPH oxidase-2/4 (NOX-2/4) expressions in cardiomyocytes. 3-methylquercetin 36-48 heme oxygenase 1 Homo sapiens 186-190 29730426-7 2018 However, SIRT1 inhibition (Sirtinol) not only inhibited isorhamnetin-induced Nrf2/HO-1 upregulation and NOX-2/4 downregulation, but also alleviated its anti-apoptotic effects. sirtinol 27-35 heme oxygenase 1 Homo sapiens 82-86 29730426-7 2018 However, SIRT1 inhibition (Sirtinol) not only inhibited isorhamnetin-induced Nrf2/HO-1 upregulation and NOX-2/4 downregulation, but also alleviated its anti-apoptotic effects. 3-methylquercetin 56-68 heme oxygenase 1 Homo sapiens 82-86 29730426-8 2018 Taken together, these data indicate that isorhamnetin can exhibit positive effect on H/R-induced injure by attenuating apoptosis and oxidative stress in H9c2 cardiomyocytes, which is partly attributable to the upregulation of SIRT1 and Nrf2/HO-1-mediated antioxidant signaling pathway. 3-methylquercetin 41-53 heme oxygenase 1 Homo sapiens 241-245 29730426-8 2018 Taken together, these data indicate that isorhamnetin can exhibit positive effect on H/R-induced injure by attenuating apoptosis and oxidative stress in H9c2 cardiomyocytes, which is partly attributable to the upregulation of SIRT1 and Nrf2/HO-1-mediated antioxidant signaling pathway. r 87-88 heme oxygenase 1 Homo sapiens 241-245 29679944-0 2018 Epigallocatechin-3-gallate attenuates microcystin-LR-induced apoptosis in human umbilical vein endothelial cells through activation of the NRF2/HO-1 pathway. epigallocatechin gallate 0-26 heme oxygenase 1 Homo sapiens 144-148 29698685-0 2018 Effect of hemin, baicalein and heme oxygenase-1 (HO-1) enzyme activity inhibitors on Cd-induced accumulation of HO-1, HSPs and aggresome-like structures in Xenopus kidney epithelial cells. Cadmium 85-87 heme oxygenase 1 Homo sapiens 31-47 29698685-0 2018 Effect of hemin, baicalein and heme oxygenase-1 (HO-1) enzyme activity inhibitors on Cd-induced accumulation of HO-1, HSPs and aggresome-like structures in Xenopus kidney epithelial cells. Cadmium 85-87 heme oxygenase 1 Homo sapiens 49-53 29698685-0 2018 Effect of hemin, baicalein and heme oxygenase-1 (HO-1) enzyme activity inhibitors on Cd-induced accumulation of HO-1, HSPs and aggresome-like structures in Xenopus kidney epithelial cells. Cadmium 85-87 heme oxygenase 1 Homo sapiens 112-116 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Heme 40-44 heme oxygenase 1 Homo sapiens 58-62 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Iron 117-121 heme oxygenase 1 Homo sapiens 40-56 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Iron 117-121 heme oxygenase 1 Homo sapiens 58-62 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Biliverdine 126-136 heme oxygenase 1 Homo sapiens 40-56 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Biliverdine 126-136 heme oxygenase 1 Homo sapiens 58-62 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Cadmium Chloride 213-218 heme oxygenase 1 Homo sapiens 40-56 29698685-3 2018 In mammalian cells, the accumulation of heme oxygenase-1 (HO-1), which catalyzes the breakdown of heme into CO, free iron and biliverdin, was reported to protect cells against potentially lethal concentrations of CdCl2. Cadmium Chloride 213-218 heme oxygenase 1 Homo sapiens 58-62 29698685-4 2018 In the present study, CdCl2 treatment of A6 kidney epithelial cells, derived from the frog, Xenopus laevis, induced the accumulation of HO-1, heat shock protein 70 (HSP70) and HSP30 as well as an increase in the production of aggregated protein and aggresome-like structures. Cadmium Chloride 22-27 heme oxygenase 1 Homo sapiens 136-140 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. S-Nitroso-N-propionyl-D,L-penicillamine 80-84 heme oxygenase 1 Homo sapiens 38-42 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. S-Nitroso-N-propionyl-D,L-penicillamine 80-84 heme oxygenase 1 Homo sapiens 200-204 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. zinc protoporphyrin 90-109 heme oxygenase 1 Homo sapiens 38-42 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. zinc protoporphyrin 90-109 heme oxygenase 1 Homo sapiens 200-204 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. zinc protoporphyrin 111-115 heme oxygenase 1 Homo sapiens 38-42 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. Cadmium Chloride 127-132 heme oxygenase 1 Homo sapiens 38-42 29698685-5 2018 Treatment of cells with inhibitors of HO-1 enzyme activity, tin protoporphyrin (SnPP) and zinc protoporphyrin (ZnPP), enhanced CdCl2-induced actin cytoskeletal disorganization and the accumulation of HO-1, HSP70, aggregated protein and aggresome-like structures. Cadmium Chloride 127-132 heme oxygenase 1 Homo sapiens 200-204 29698685-9 2018 These results suggest that HO-1-mediated protection against CdCl2 toxicity includes the maintenance of actin cytoskeletal and microtubular structure and the suppression of aggregated protein and aggresome-like structures. Cadmium Chloride 60-65 heme oxygenase 1 Homo sapiens 27-31 29960846-4 2018 Treatment of Hep3B with BPA increased the levels of nitrous oxide, a metabolite of nitric oxide and activated Nrf2 by nitrosylation of Keap1, leading to the induction of heme oxygenase-1 (HO-1) and UGT2B1 mRNAs. bisphenol A 24-27 heme oxygenase 1 Homo sapiens 170-186 29960846-4 2018 Treatment of Hep3B with BPA increased the levels of nitrous oxide, a metabolite of nitric oxide and activated Nrf2 by nitrosylation of Keap1, leading to the induction of heme oxygenase-1 (HO-1) and UGT2B1 mRNAs. Nitrous Oxide 52-65 heme oxygenase 1 Homo sapiens 170-186 29679944-8 2018 Our findings indicate that EGCG treatment protects against MC-LR-mediated HUVEC apoptosis via activation of NRF2/HO-1 signaling. epigallocatechin gallate 27-31 heme oxygenase 1 Homo sapiens 113-117 29679944-8 2018 Our findings indicate that EGCG treatment protects against MC-LR-mediated HUVEC apoptosis via activation of NRF2/HO-1 signaling. cyanoginosin LR 59-64 heme oxygenase 1 Homo sapiens 113-117 29679944-7 2018 EGCG induced NRF2/HO-1 expression and activation in MC-LR treated HUVECs, while downregulation of NRF2/HO-1 by specific siRNAs revealed that NRF2/HO-1 signaling was involved in EGCG attenuation of MC-LR-induced HUVEC apoptosis. epigallocatechin gallate 0-4 heme oxygenase 1 Homo sapiens 18-22 29679944-7 2018 EGCG induced NRF2/HO-1 expression and activation in MC-LR treated HUVECs, while downregulation of NRF2/HO-1 by specific siRNAs revealed that NRF2/HO-1 signaling was involved in EGCG attenuation of MC-LR-induced HUVEC apoptosis. cyanoginosin LR 52-57 heme oxygenase 1 Homo sapiens 18-22 29753142-0 2018 Heme Oxygenase-1 inhibits spring viremia of carp virus replication through carbon monoxide mediated cyclic GMP/Protein kinase G signaling pathway. Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 0-16 29753142-0 2018 Heme Oxygenase-1 inhibits spring viremia of carp virus replication through carbon monoxide mediated cyclic GMP/Protein kinase G signaling pathway. Cyclic GMP 100-110 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Heme 69-73 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Heme 69-73 heme oxygenase 1 Homo sapiens 18-22 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Carbon Monoxide 85-100 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Carbon Monoxide 85-100 heme oxygenase 1 Homo sapiens 18-22 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Carbon Monoxide 102-104 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Carbon Monoxide 102-104 heme oxygenase 1 Homo sapiens 18-22 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Biliverdine 107-117 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Biliverdine 107-117 heme oxygenase 1 Homo sapiens 18-22 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Iron 130-134 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. Iron 130-134 heme oxygenase 1 Homo sapiens 18-22 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. ammonium ferrous sulfate 136-140 heme oxygenase 1 Homo sapiens 0-16 29753142-3 2018 Heme oxygenase-1 (HO-1), an enzyme that catalyzes the degradation of heme to produce carbon monoxide (CO), biliverdin and ferrous iron (Fe2+), exerts anti-oxidant, antiinflammatory and anti-apoptotic properties. ammonium ferrous sulfate 136-140 heme oxygenase 1 Homo sapiens 18-22 29753142-6 2018 The elevated expression of HO-1 was induced upon cobalt protoporphyrin (CoPP) treatment in EPC cells without affecting cell viability and thus inhibited SVCV replication in a dose dependent manner. cobaltiprotoporphyrin 49-70 heme oxygenase 1 Homo sapiens 27-31 29753142-6 2018 The elevated expression of HO-1 was induced upon cobalt protoporphyrin (CoPP) treatment in EPC cells without affecting cell viability and thus inhibited SVCV replication in a dose dependent manner. cobaltiprotoporphyrin 72-76 heme oxygenase 1 Homo sapiens 27-31 29753142-8 2018 Thereby, the antiviral activity of ROS/Nrf2/HO-1 axis was confirmed in EPC cells. ros 35-38 heme oxygenase 1 Homo sapiens 44-48 29753142-10 2018 Collectively, these findings suggest potential drug or therapy that induced the Nrf2/HO-1/CO/cGMP/PKG signaling pathway as a promising strategy for treating SVC. Cyclic GMP 93-97 heme oxygenase 1 Homo sapiens 85-92 29939160-4 2018 Noncanonical ferroptosis induction is characterized by an increase in intracellular labile Fe(II) upon excessive activation of heme oxygenase-1, which is sufficient to induce ferroptosis. ammonium ferrous sulfate 91-97 heme oxygenase 1 Homo sapiens 127-143 29906742-10 2018 The expression of Nrf2 and HO-1 were dose-dependently up-regulated by the treatment of xanthohumol. xanthohumol 87-98 heme oxygenase 1 Homo sapiens 27-31 29909234-6 2018 Additionally, the Puerarin treatment also significantly enhanced (P < 0.001) the mRNA expressions levels of the anti-oxidant enzymes such as Nrf2, HO-1 and SOD2. puerarin 18-26 heme oxygenase 1 Homo sapiens 150-154 31949812-0 2018 Overexpression of miR-153 promotes oxidative stress in MPP+-induced PD model by negatively regulating the Nrf2/HO-1 signaling pathway. 1-(4-methoxyphenyl)pyridinium 55-59 heme oxygenase 1 Homo sapiens 111-115 31949812-10 2018 Finally, it was observed that miR-153 could suppress the Nrf2/HO-1 signaling pathway in MPP+-treated SH-SY5Y cells. 1-(4-methoxyphenyl)pyridinium 88-91 heme oxygenase 1 Homo sapiens 62-66 30106049-8 2018 Compared with the control group, the expression of NFE2L2 and HMOX1 was significantly increased in the genistein + rotenone group. Genistein 103-112 heme oxygenase 1 Homo sapiens 62-67 29473125-0 2018 Sulforaphane Attenuated the Pro-Inflammatory State Induced by Hydrogen Peroxide in SH-SY5Y Cells Through the Nrf2/HO-1 Signaling Pathway. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 114-118 29473125-0 2018 Sulforaphane Attenuated the Pro-Inflammatory State Induced by Hydrogen Peroxide in SH-SY5Y Cells Through the Nrf2/HO-1 Signaling Pathway. Hydrogen Peroxide 62-79 heme oxygenase 1 Homo sapiens 114-118 29473125-8 2018 Therefore, SFN exerted an anti-inflammatory effect in H2O2-challenged SH-SY5Y cells by a mechanism dependent on the Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 54-58 heme oxygenase 1 Homo sapiens 121-125 30106049-8 2018 Compared with the control group, the expression of NFE2L2 and HMOX1 was significantly increased in the genistein + rotenone group. Rotenone 115-123 heme oxygenase 1 Homo sapiens 62-67 30187882-5 2018 In HepG2 cells, treatment with triptolide significantly inhibited the cell growth, resulting in a cell viability as low as 72.83% at 24 h; triptolide also induced obvious cell apoptosis and cell nucleus deformation, causing an apoptosis rate of 43.1% in the cells at 24 h. Triptolide significantly reduced the expressions of Nrf2 and HO-1 proteins related with the oxidative stress pathway. triptolide 31-41 heme oxygenase 1 Homo sapiens 334-338 29732634-4 2018 Well-known pathways such as Pl3K/Akt/Bcl-2 pathway, Nrf2/HO-1 pathway, and MAPK signaling pathway help berberine to protect neurons through antiapoptotic, antioxidative, and anti-inflammatory activities. Berberine 103-112 heme oxygenase 1 Homo sapiens 57-61 30060630-16 2018 Moreover, the expression of HO-1 and NQO-1 increased significantly in the CTP group. Cytidine Triphosphate 74-77 heme oxygenase 1 Homo sapiens 28-32 30084803-0 2018 Pyridoxine exerts antioxidant effects in cell model of Alzheimer"s disease via the Nrf-2/HO-1 pathway. Pyridoxine 0-10 heme oxygenase 1 Homo sapiens 89-93 30084803-5 2018 The results obtained revealed that pyridoxine exerted a protective potential against AD, attenuated ROS levels, decreased the expressions of cytoplasmic Nrf2, and upregulated whole-cell HO-1 expression. Pyridoxine 35-45 heme oxygenase 1 Homo sapiens 186-190 30084803-6 2018 These results suggest that the anti-AD effect of pyridoxine may be attributed to its anti-oxidant property elicited via stimulation of the Nrf2/HO-1 pathway. Pyridoxine 49-59 heme oxygenase 1 Homo sapiens 144-148 30187882-5 2018 In HepG2 cells, treatment with triptolide significantly inhibited the cell growth, resulting in a cell viability as low as 72.83% at 24 h; triptolide also induced obvious cell apoptosis and cell nucleus deformation, causing an apoptosis rate of 43.1% in the cells at 24 h. Triptolide significantly reduced the expressions of Nrf2 and HO-1 proteins related with the oxidative stress pathway. triptolide 139-149 heme oxygenase 1 Homo sapiens 334-338 29678719-0 2018 Carnosic acid attenuates cartilage degeneration through induction of heme oxygenase-1 in human articular chondrocytes. salvin 0-13 heme oxygenase 1 Homo sapiens 69-85 30048241-4 2018 METHODS AND RESULTS: Upregulation of HO-1 by Hemin or adenovirus infection reversed H2O2-induced senescence in human umbilical vein endothelial cells (HUVECs); whereas depletion of HO-1 by siRNA or HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) triggered HUVEC senescence. Hydrogen Peroxide 84-88 heme oxygenase 1 Homo sapiens 37-41 30048241-4 2018 METHODS AND RESULTS: Upregulation of HO-1 by Hemin or adenovirus infection reversed H2O2-induced senescence in human umbilical vein endothelial cells (HUVECs); whereas depletion of HO-1 by siRNA or HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) triggered HUVEC senescence. protoporphyrin IX 213-230 heme oxygenase 1 Homo sapiens 37-41 30048241-4 2018 METHODS AND RESULTS: Upregulation of HO-1 by Hemin or adenovirus infection reversed H2O2-induced senescence in human umbilical vein endothelial cells (HUVECs); whereas depletion of HO-1 by siRNA or HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) triggered HUVEC senescence. zinc protoporphyrin 242-246 heme oxygenase 1 Homo sapiens 37-41 30048241-5 2018 Mechanistically, overexpression of HO-1 enhanced the interaction between HO-1 and endothelial nitric oxide synthase (eNOS), and promoted the interaction between eNOS and its upstream kinase Akt, thus resulting in an enhancement of eNOS phosphorylation at Ser1177 and a subsequent increase of nitric oxide (NO) production. Nitric Oxide 94-106 heme oxygenase 1 Homo sapiens 35-39 30048241-5 2018 Mechanistically, overexpression of HO-1 enhanced the interaction between HO-1 and endothelial nitric oxide synthase (eNOS), and promoted the interaction between eNOS and its upstream kinase Akt, thus resulting in an enhancement of eNOS phosphorylation at Ser1177 and a subsequent increase of nitric oxide (NO) production. Nitric Oxide 94-106 heme oxygenase 1 Homo sapiens 73-77 30048241-6 2018 Moreover, HO-1 induction prevented the decrease of eNOS dimer/monomer ratio stimulated by H2O2 via its antioxidant properties. Hydrogen Peroxide 90-94 heme oxygenase 1 Homo sapiens 10-14 29980703-3 2018 The plant-derived polyphenols, carnosol and curcumin, have been identified as candidate HO-1 inducers however there has been little investigation into their effects on human immune cells. Polyphenols 18-29 heme oxygenase 1 Homo sapiens 88-92 29980703-3 2018 The plant-derived polyphenols, carnosol and curcumin, have been identified as candidate HO-1 inducers however there has been little investigation into their effects on human immune cells. carnosol 31-39 heme oxygenase 1 Homo sapiens 88-92 29980703-3 2018 The plant-derived polyphenols, carnosol and curcumin, have been identified as candidate HO-1 inducers however there has been little investigation into their effects on human immune cells. Curcumin 44-52 heme oxygenase 1 Homo sapiens 88-92 30097118-10 2018 Hypericin reduced HO-1 and NS5A in a time- and dose- dependent manner. hypericin 0-9 heme oxygenase 1 Homo sapiens 18-22 30097118-11 2018 Chidamide, but not 5-Aza-dc, restored hypericin-induced reduction in HCV NS3 expression and reversed HO-1 expression in Ava5 cells. N-(2-amino-5-fluorobenzyl)-4-(N-(pyridine-3-acrylyl)aminomethyl)benzamide 0-9 heme oxygenase 1 Homo sapiens 101-105 30097118-12 2018 LY294002 inhibited HCV replication via HO-1 down-regulation. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 heme oxygenase 1 Homo sapiens 39-43 30097118-15 2018 CONCLUSION: In conclusion, hypericin inhibits HCV replication via down-regulation of HO-1 expression and deacetylation. hypericin 27-36 heme oxygenase 1 Homo sapiens 85-89 29792861-7 2018 Conversely, Akt-mTORC1 inhibitors (RAD001 and LY294002) reduced NLGN3-induced HO1, NOQ1 and GCLC mRNA expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 46-54 heme oxygenase 1 Homo sapiens 78-81 29678719-5 2018 Among the signaling pathways that control cellular protective mechanisms against oxygen radical damage is heme oxygenase-1 (HO-1). Oxygen 81-87 heme oxygenase 1 Homo sapiens 106-122 29678719-5 2018 Among the signaling pathways that control cellular protective mechanisms against oxygen radical damage is heme oxygenase-1 (HO-1). Oxygen 81-87 heme oxygenase 1 Homo sapiens 124-128 29678719-8 2018 Carnosic acid (CA), a natural diterpene with oxidant activity, is prevents cartilage degeneration though induction of HO-1. salvin 0-13 heme oxygenase 1 Homo sapiens 118-122 29452096-1 2018 Heme oxygenase (HO)-1, the inducible isoform of the heme-degrading enzyme HO, plays a critical role in inflammation and iron homeostasis. Heme 52-56 heme oxygenase 1 Homo sapiens 0-21 30057686-0 2018 Flutamide Induces Hepatic Cell Death and Mitochondrial Dysfunction via Inhibition of Nrf2-Mediated Heme Oxygenase-1. Flutamide 0-9 heme oxygenase 1 Homo sapiens 99-115 30057686-4 2018 This study was designed to delineate the role of Nrf2/HO-1 in flutamide-induced hepatic cell injury. Flutamide 62-71 heme oxygenase 1 Homo sapiens 54-58 30057686-6 2018 The protein expression of Nrf2 and HO-1 was induced by flutamide at 12.5 muM but was downregulated by higher concentrations of flutamide. Flutamide 55-64 heme oxygenase 1 Homo sapiens 35-39 30057686-6 2018 The protein expression of Nrf2 and HO-1 was induced by flutamide at 12.5 muM but was downregulated by higher concentrations of flutamide. Flutamide 127-136 heme oxygenase 1 Homo sapiens 35-39 30057686-7 2018 Silencing either Nrf2 or HO-1 was found to aggravate flutamide-induced hydrogen peroxide accumulation and mitochondrial dysfunction as well as inhibition of the Nrf2 pathway. Flutamide 53-62 heme oxygenase 1 Homo sapiens 25-29 30057686-7 2018 Silencing either Nrf2 or HO-1 was found to aggravate flutamide-induced hydrogen peroxide accumulation and mitochondrial dysfunction as well as inhibition of the Nrf2 pathway. Hydrogen Peroxide 71-88 heme oxygenase 1 Homo sapiens 25-29 30057686-8 2018 Moreover, preinduction of HO-1 by Copp significantly attenuated flutamide-induced oxidative stress and mitochondrial dysfunction, while inhibition of HO-1 by Snpp aggravated these deleterious effects. COPP protocol 34-38 heme oxygenase 1 Homo sapiens 26-30 30057686-8 2018 Moreover, preinduction of HO-1 by Copp significantly attenuated flutamide-induced oxidative stress and mitochondrial dysfunction, while inhibition of HO-1 by Snpp aggravated these deleterious effects. Flutamide 64-73 heme oxygenase 1 Homo sapiens 26-30 30057686-9 2018 These findings suggest that flutamide-induced hepatic cell death and mitochondrial dysfunction is assoicated with inhibition of Nrf2-mediated HO-1. Flutamide 28-37 heme oxygenase 1 Homo sapiens 142-146 30057686-10 2018 Pharmacologic intervention of Nrf2/HO-1 may provide a promising therapeutic approach in flutamide-induced liver injury. Flutamide 88-97 heme oxygenase 1 Homo sapiens 35-39 29452096-1 2018 Heme oxygenase (HO)-1, the inducible isoform of the heme-degrading enzyme HO, plays a critical role in inflammation and iron homeostasis. Iron 120-124 heme oxygenase 1 Homo sapiens 0-21 29656184-0 2018 Ropivacaine regulates the expression and function of heme oxygenase-1. Ropivacaine 0-11 heme oxygenase 1 Homo sapiens 53-69 29582209-7 2018 Increased ROS levels were also observed, possibly attributed to the weakened anti-oxidative response evidenced by the underexpression of the Nrf2-regulated genes encoding HO-1 and NQO1. Reactive Oxygen Species 10-13 heme oxygenase 1 Homo sapiens 171-175 29656184-4 2018 In the current study, our results indicated that ropivacaine treatment caused a significant induction of heme oxygenase-1 (HO-1) at both the mRNA and protein levels in human SHSY5Y cells. Ropivacaine 49-60 heme oxygenase 1 Homo sapiens 105-121 29656184-6 2018 Additionally, ropivacaine treatment enhanced HO-1 activity in a dose-dependent manner. Ropivacaine 14-25 heme oxygenase 1 Homo sapiens 45-49 29656184-4 2018 In the current study, our results indicated that ropivacaine treatment caused a significant induction of heme oxygenase-1 (HO-1) at both the mRNA and protein levels in human SHSY5Y cells. Ropivacaine 49-60 heme oxygenase 1 Homo sapiens 123-127 29656184-8 2018 Blockage of p38 phosphorylation with its specific inhibitor SB203580 or by transfection with p38 siRNA restrained ropivacaine-stimulated HO-1 expression. SB 203580 60-68 heme oxygenase 1 Homo sapiens 137-141 29656184-8 2018 Blockage of p38 phosphorylation with its specific inhibitor SB203580 or by transfection with p38 siRNA restrained ropivacaine-stimulated HO-1 expression. Ropivacaine 114-125 heme oxygenase 1 Homo sapiens 137-141 29656184-10 2018 Silencing of Nrf2 abolished ropivacaine-induced HO-1 expression. Ropivacaine 28-39 heme oxygenase 1 Homo sapiens 48-52 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Ropivacaine 60-71 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Ropivacaine 60-71 heme oxygenase 1 Homo sapiens 237-241 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Reactive Oxygen Species 94-117 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Reactive Oxygen Species 119-122 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Glutathione 145-156 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Glutathione 158-161 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Ropivacaine 245-256 heme oxygenase 1 Homo sapiens 37-41 29656184-11 2018 Notably, we found that inhibition of HO-1 activity promoted ropivacaine-induced production of reactive oxygen species (ROS), deletion of reduced glutathione (GSH), and release of lactate dehydrogenase (LDH), suggesting that induction of HO-1 by ropivacaine acted as a compensatory survival response against ropivacaine. Ropivacaine 245-256 heme oxygenase 1 Homo sapiens 37-41 29656184-5 2018 Levels of HO-1 mRNA and protein peaked at 1 h and 18 h, respectively, in response to ropivacaine treatment. Ropivacaine 85-96 heme oxygenase 1 Homo sapiens 10-14 29758489-8 2018 Furthermore, fisetin significantly increased the effects of the protective antioxidant pathway by promoting the expression of nuclear factor erythroid-2-related factor-2 and heme oxygenase 1. fisetin 13-20 heme oxygenase 1 Homo sapiens 174-190 29693710-3 2018 HO-1, by regulating intracellular levels of pro-oxidant heme, or by other benefits of its by-products such as carbon monoxide (CO) and biliverdin (BV) had become an important candidate protein to be up-regulated to combat diverse stressful events. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 0-4 29896252-4 2018 HSCs-T6 were incubated with different concentrations of hemin (HO-1 chemical inducer) and Znpp-IX (HO-1 chemical inhibitor) for 12, 24 and 48 h. Cell viability was determined using an MTT assay. Hemin 56-61 heme oxygenase 1 Homo sapiens 63-67 29896252-4 2018 HSCs-T6 were incubated with different concentrations of hemin (HO-1 chemical inducer) and Znpp-IX (HO-1 chemical inhibitor) for 12, 24 and 48 h. Cell viability was determined using an MTT assay. zinc protoporphyrin 90-97 heme oxygenase 1 Homo sapiens 99-103 29751106-6 2018 Curcumin significantly induced HMOX1 in single cell type models and co-cultures. Curcumin 0-8 heme oxygenase 1 Homo sapiens 31-36 29693114-0 2018 Human circular RNA-0054633 regulates high glucose-induced vascular endothelial cell dysfunction through the microRNA-218/roundabout 1 and microRNA-218/heme oxygenase-1 axes. Glucose 42-49 heme oxygenase 1 Homo sapiens 138-167 29693114-9 2018 It was also demonstrated that downregulating the expression of miR-218 inhibited the high glucose-induced endothelial cell dysfunction by promoting the expression of ROBO1 and HO-1. Glucose 90-97 heme oxygenase 1 Homo sapiens 176-180 29693710-3 2018 HO-1, by regulating intracellular levels of pro-oxidant heme, or by other benefits of its by-products such as carbon monoxide (CO) and biliverdin (BV) had become an important candidate protein to be up-regulated to combat diverse stressful events. Carbon Monoxide 127-129 heme oxygenase 1 Homo sapiens 0-4 29693710-3 2018 HO-1, by regulating intracellular levels of pro-oxidant heme, or by other benefits of its by-products such as carbon monoxide (CO) and biliverdin (BV) had become an important candidate protein to be up-regulated to combat diverse stressful events. Biliverdine 135-145 heme oxygenase 1 Homo sapiens 0-4 29693710-3 2018 HO-1, by regulating intracellular levels of pro-oxidant heme, or by other benefits of its by-products such as carbon monoxide (CO) and biliverdin (BV) had become an important candidate protein to be up-regulated to combat diverse stressful events. Biliverdine 147-149 heme oxygenase 1 Homo sapiens 0-4 29498764-8 2018 Moreover, we showed that both in animal model and in human liver, frequent hemin infusions generate a chronic inflammatory hepatic disease which induces HO1 remotely to hemin treatment and maintains a high ALAS1 level responsible for recurrence. Hemin 75-80 heme oxygenase 1 Homo sapiens 153-156 29498764-8 2018 Moreover, we showed that both in animal model and in human liver, frequent hemin infusions generate a chronic inflammatory hepatic disease which induces HO1 remotely to hemin treatment and maintains a high ALAS1 level responsible for recurrence. Hemin 169-174 heme oxygenase 1 Homo sapiens 153-156 29704670-7 2018 The expression of Nrf2 and HO-1 were up-regulated by treatment of isorhamnetin. 3-methylquercetin 66-78 heme oxygenase 1 Homo sapiens 27-31 29951371-6 2018 In addition, endogenous miR-494 inhibition in undifferentiated cells impairs HO-1 induction in response to exposure to 500 microM H2O2, reducing the number of viable cells. Hydrogen Peroxide 130-134 heme oxygenase 1 Homo sapiens 77-81 29997086-8 2018 CONCLUSION: Agmatine inhibits LPS-induced activation and dysfunction of HUVECs by modulating NF-kappaB and MAPK signal pathways to down-regulate the expressions of adhesion molecules and chemokines and by up-regulating the expression of HO-1 to reduce ROS production. Agmatine 12-20 heme oxygenase 1 Homo sapiens 237-241 29997086-7 2018 Agmatine significantly up-regulated the mRNA expression of HO-1 (P<0.05), inhibited LPS-induced phosphorylation of p38, ERK, nuclear p65 and cytoplasmic IkappaBalpha, and up-regulated the protein expression of cytoplasmic IkappaBalpha. Agmatine 0-8 heme oxygenase 1 Homo sapiens 59-63 29921869-9 2018 Iron deposition associated with oxidative injury as indicated by heme oxygenase-1 abundance. Iron 0-4 heme oxygenase 1 Homo sapiens 65-81 29951371-7 2018 The analysis of Bach1 expression did not reveal any significant modifications in any experimental conditions tested, proving that the impairment of HO-1 induction observed in cells treated with miR-494 inhibitor and exposed to H2O2 is independent from Bach1. Hydrogen Peroxide 227-231 heme oxygenase 1 Homo sapiens 148-152 29704732-4 2018 Furthermore, paeonol treatment resulted in the increase of Nrf2 nuclear translocation, the increase of NQO-1 and HO-1 expression, and the suppression of NF-kappaB p65 nuclear translocation. paeonol 13-20 heme oxygenase 1 Homo sapiens 113-117 29874784-8 2018 In addition, EESS increased not only expression, but also phosphorylation of nuclear factor-erythroid 2 related factor 2 (Nrf2), and promoted the expression of heme oxygenase-1 (HO-1), a critical target enzyme of Nrf2, but decreased the expression of kelch-like ECH-associated protein-1; however, the inhibition of HO-1 activity by zinc protoporphyrin abolished the antioxidant potential induced by EESS against H2O2-mediated oxidative stress. zinc protoporphyrin 332-351 heme oxygenase 1 Homo sapiens 160-176 29874784-8 2018 In addition, EESS increased not only expression, but also phosphorylation of nuclear factor-erythroid 2 related factor 2 (Nrf2), and promoted the expression of heme oxygenase-1 (HO-1), a critical target enzyme of Nrf2, but decreased the expression of kelch-like ECH-associated protein-1; however, the inhibition of HO-1 activity by zinc protoporphyrin abolished the antioxidant potential induced by EESS against H2O2-mediated oxidative stress. zinc protoporphyrin 332-351 heme oxygenase 1 Homo sapiens 178-182 29874784-8 2018 In addition, EESS increased not only expression, but also phosphorylation of nuclear factor-erythroid 2 related factor 2 (Nrf2), and promoted the expression of heme oxygenase-1 (HO-1), a critical target enzyme of Nrf2, but decreased the expression of kelch-like ECH-associated protein-1; however, the inhibition of HO-1 activity by zinc protoporphyrin abolished the antioxidant potential induced by EESS against H2O2-mediated oxidative stress. Hydrogen Peroxide 412-416 heme oxygenase 1 Homo sapiens 160-176 29874784-8 2018 In addition, EESS increased not only expression, but also phosphorylation of nuclear factor-erythroid 2 related factor 2 (Nrf2), and promoted the expression of heme oxygenase-1 (HO-1), a critical target enzyme of Nrf2, but decreased the expression of kelch-like ECH-associated protein-1; however, the inhibition of HO-1 activity by zinc protoporphyrin abolished the antioxidant potential induced by EESS against H2O2-mediated oxidative stress. Hydrogen Peroxide 412-416 heme oxygenase 1 Homo sapiens 178-182 29626606-6 2018 Moreover, curcumin markedly enhanced the translocation of Nrf2 from the cytoplasm to the nucleus, proved by the results of western blot and immunofluorescence, subsequently increased the expression of downstream factors such as heme oxygenase 1 (HO1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) and prevented the decline of antioxidant enzyme activities. Curcumin 10-18 heme oxygenase 1 Homo sapiens 228-244 29626606-6 2018 Moreover, curcumin markedly enhanced the translocation of Nrf2 from the cytoplasm to the nucleus, proved by the results of western blot and immunofluorescence, subsequently increased the expression of downstream factors such as heme oxygenase 1 (HO1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) and prevented the decline of antioxidant enzyme activities. Curcumin 10-18 heme oxygenase 1 Homo sapiens 246-249 29704732-5 2018 However, pretreatment with NAM (inhibitor of SIRT1) not only inhibited the effect of paeonol on reducing nuclear translocation of NF-kappaBp65, but also inhibited the effect of paeonol on promoting the expression of nuclear Nrf2, NQO1 and HO-1. paeonol 85-92 heme oxygenase 1 Homo sapiens 239-243 29704732-5 2018 However, pretreatment with NAM (inhibitor of SIRT1) not only inhibited the effect of paeonol on reducing nuclear translocation of NF-kappaBp65, but also inhibited the effect of paeonol on promoting the expression of nuclear Nrf2, NQO1 and HO-1. paeonol 177-184 heme oxygenase 1 Homo sapiens 239-243 29477863-7 2018 These divergences in ROS production seem to be correlated with the different extension of intracellular signaling pathways activation and, by consequence, with distinct transcription kinetics of genes such as HMOX1, IL8, IL1B and CD86. Reactive Oxygen Species 21-24 heme oxygenase 1 Homo sapiens 209-214 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Heme 96-100 heme oxygenase 1 Homo sapiens 0-16 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Heme 96-100 heme oxygenase 1 Homo sapiens 18-22 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 0-16 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 18-22 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Iron 123-127 heme oxygenase 1 Homo sapiens 0-16 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Iron 123-127 heme oxygenase 1 Homo sapiens 18-22 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Carbon Monoxide 133-148 heme oxygenase 1 Homo sapiens 0-16 29772541-2 2018 Heme oxygenase-1 (HO-1), a microsomal enzyme discovered decades ago, can metabolize pro-oxidant heme into biliverdin, free iron, and carbon monoxide. Carbon Monoxide 133-148 heme oxygenase 1 Homo sapiens 18-22 29532858-8 2018 All of the results indicated that hydrogen decreased the levels of reactive oxygen species, 8-iso-prostaglandin F2alpha and malondialdehyde, and promoted the UVB exposure-induced expression of PI3K, Akt, Nrf2 and heme oxygenase-1 in HaCaT cells. Hydrogen 34-42 heme oxygenase 1 Homo sapiens 213-229 29532858-10 2018 Therefore, hydrogen effectively protects cells from UVB radiation-induced oxidative stress by inhibiting Nrf2/HO-1 activation through the PI3K/Akt signaling pathway. Hydrogen 11-19 heme oxygenase 1 Homo sapiens 110-114 29194609-10 2018 Based on gene expression data of withaferin-A and salbutamol, we identified HMOX1 and STC1 as being strongly differentially expressed . withaferin A 33-45 heme oxygenase 1 Homo sapiens 76-81 29194609-10 2018 Based on gene expression data of withaferin-A and salbutamol, we identified HMOX1 and STC1 as being strongly differentially expressed . Albuterol 50-60 heme oxygenase 1 Homo sapiens 76-81 29668263-7 2018 In addition, ALA treatment inhibited reactive oxygen species generation induced by Abeta25-35 through the enhancement of the nuclear factor-erythroid 2-related factor-2 (Nrf-2) protein levels and subsequent induction of heme-oxygenase-1 (HO-1) expression in C6 glial cells dose- and time-dependently. alpha-Linolenic Acid 13-16 heme oxygenase 1 Homo sapiens 220-236 29783634-0 2018 Novel Structural Insight into Inhibitors of Heme Oxygenase-1 (HO-1) by New Imidazole-Based Compounds: Biochemical and In Vitro Anticancer Activity Evaluation. imidazole 75-84 heme oxygenase 1 Homo sapiens 44-60 29783634-0 2018 Novel Structural Insight into Inhibitors of Heme Oxygenase-1 (HO-1) by New Imidazole-Based Compounds: Biochemical and In Vitro Anticancer Activity Evaluation. imidazole 75-84 heme oxygenase 1 Homo sapiens 62-66 29783634-1 2018 In this paper, the design, synthesis, and molecular modeling of a new azole-based HO-1 inhibitors was reported, using compound 1 as a lead compound, in which an imidazole moiety is linked to a hydrophobic group by means of an ethanolic spacer. Azoles 70-75 heme oxygenase 1 Homo sapiens 82-86 29783634-1 2018 In this paper, the design, synthesis, and molecular modeling of a new azole-based HO-1 inhibitors was reported, using compound 1 as a lead compound, in which an imidazole moiety is linked to a hydrophobic group by means of an ethanolic spacer. imidazole 161-170 heme oxygenase 1 Homo sapiens 82-86 29783634-4 2018 Molecular modeling studies confirmed a consolidated binding mode in which the nitrogen of the imidazolyl moiety coordinated the heme ferrous iron, meanwhile the hydrophobic groups were located in the western region of HO-1 binding pocket. Nitrogen 78-86 heme oxygenase 1 Homo sapiens 218-222 29783634-4 2018 Molecular modeling studies confirmed a consolidated binding mode in which the nitrogen of the imidazolyl moiety coordinated the heme ferrous iron, meanwhile the hydrophobic groups were located in the western region of HO-1 binding pocket. imidazolyl 94-104 heme oxygenase 1 Homo sapiens 218-222 29783634-4 2018 Molecular modeling studies confirmed a consolidated binding mode in which the nitrogen of the imidazolyl moiety coordinated the heme ferrous iron, meanwhile the hydrophobic groups were located in the western region of HO-1 binding pocket. Heme 128-132 heme oxygenase 1 Homo sapiens 218-222 29783634-4 2018 Molecular modeling studies confirmed a consolidated binding mode in which the nitrogen of the imidazolyl moiety coordinated the heme ferrous iron, meanwhile the hydrophobic groups were located in the western region of HO-1 binding pocket. Iron 141-145 heme oxygenase 1 Homo sapiens 218-222 29668263-7 2018 In addition, ALA treatment inhibited reactive oxygen species generation induced by Abeta25-35 through the enhancement of the nuclear factor-erythroid 2-related factor-2 (Nrf-2) protein levels and subsequent induction of heme-oxygenase-1 (HO-1) expression in C6 glial cells dose- and time-dependently. alpha-Linolenic Acid 13-16 heme oxygenase 1 Homo sapiens 238-242 29668263-9 2018 In conclusion, effects of ALA on Abeta degradation were shown to be mediated through inhibition of inflammatory responses and activation of antioxidative system, Nrf-2/HO-1 signaling pathway, in C6 glial cells. alpha-Linolenic Acid 26-29 heme oxygenase 1 Homo sapiens 168-172 29491190-9 2018 Results: Our results demonstrate that pre-treatment and post-treatment with glutamine promoted melanocyte viability, increased levels of superoxide dismutase, glutathione-S-transferase and bcl-2, decreased levels of reactive oxygen species, malondialdehyde, bax and caspase-3, and enhanced nuclear factor-E2-related factor 2, heme oxygenase-1, and heat shock protein 70 expression in a dose dependent manner. Glutamine 76-85 heme oxygenase 1 Homo sapiens 298-342 29461260-5 2018 Heme may increase host susceptibility to infections by inducing heme oxygenase 1 (HO-1) in immature neutrophils, thereby inhibiting oxidative burst required for clearance of engulfed bacteria. Heme 0-4 heme oxygenase 1 Homo sapiens 64-80 29461260-5 2018 Heme may increase host susceptibility to infections by inducing heme oxygenase 1 (HO-1) in immature neutrophils, thereby inhibiting oxidative burst required for clearance of engulfed bacteria. Heme 0-4 heme oxygenase 1 Homo sapiens 82-86 29644357-5 2018 Furthermore, MH suppressed the Nrf2-dependent antioxidant enzyme expression (superoxide dismutase (SOD), catalase and heme oxygenase-1) and the activity of SOD, catalase, glutathione peroxidase and glutathione reductase. mh 13-15 heme oxygenase 1 Homo sapiens 118-134 28989083-14 2018 Furthermore, hNSCs co-treated with resveratrol were significantly rescued from Abeta-induced oxidative stress, which correlated with reversal of the Abeta-induced mRNA decrease in oxidative defense genes (SOD-1, NRF2, Gpx1, Catalase, GSH and HO-1). Resveratrol 35-46 heme oxygenase 1 Homo sapiens 242-246 29356228-0 2018 Curcuma sp.-derived dehydrocurdione induces heme oxygenase-1 through a Michael reaction between its alpha, beta-unsaturated carbonyl and Keap1. curcuma sp. 0-11 heme oxygenase 1 Homo sapiens 44-60 29356228-0 2018 Curcuma sp.-derived dehydrocurdione induces heme oxygenase-1 through a Michael reaction between its alpha, beta-unsaturated carbonyl and Keap1. dehydrocurdione 20-35 heme oxygenase 1 Homo sapiens 44-60 29356228-1 2018 To elucidate the anti-inflammatory mechanism of Curcuma sp., we investigated whether dehydrocurdione, a sesquiterpene contained in Curcuma sp., induces heme oxygenase (HO)-1, an antioxidative enzyme, in RAW 264.7 macrophages. dehydrocurdione 85-100 heme oxygenase 1 Homo sapiens 152-173 29288927-9 2018 Moreover, ZYZ-803 upregulated the endogenous antioxidants, glutathione peroxidase (GPx) and heme oxygenase 1 (HO-1). zyz-803 10-17 heme oxygenase 1 Homo sapiens 92-108 29525432-0 2018 Marliolide inhibits skin carcinogenesis by activating NRF2/ARE to induce heme oxygenase-1. marliolide 0-10 heme oxygenase 1 Homo sapiens 73-89 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Heme 63-67 heme oxygenase 1 Homo sapiens 0-16 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Heme 63-67 heme oxygenase 1 Homo sapiens 18-22 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 0-16 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 18-22 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Carbon Monoxide 126-128 heme oxygenase 1 Homo sapiens 0-16 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Carbon Monoxide 126-128 heme oxygenase 1 Homo sapiens 18-22 29568961-10 2018 Furthermore, pretreatment with diosmetin elevated mRNA and protein expression levels of Nrf2, HO-1 and NQO1. diosmetin 31-40 heme oxygenase 1 Homo sapiens 94-98 29568961-11 2018 The present study is the first, to the best of our knowledge, to demonstrate that activation of the Nrf2/NQO1-HO-1 signaling pathway maybe involved in the cytoprotective effects of diosmetin against oxidative stress. diosmetin 181-190 heme oxygenase 1 Homo sapiens 110-114 29796178-0 2018 SB202190 inhibits endothelial cell apoptosis via induction of autophagy and heme oxygenase-1. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 0-8 heme oxygenase 1 Homo sapiens 76-92 29796178-2 2018 The present study therefore investigates the impact of the p38 MAPK inhibitor SB202190 on the expression of the cytoprotective enzyme heme oxygenase-1 (HO-1) as well as metabolic activity, apoptosis and autophagy of endothelial cells. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 78-86 heme oxygenase 1 Homo sapiens 134-150 29796178-2 2018 The present study therefore investigates the impact of the p38 MAPK inhibitor SB202190 on the expression of the cytoprotective enzyme heme oxygenase-1 (HO-1) as well as metabolic activity, apoptosis and autophagy of endothelial cells. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 78-86 heme oxygenase 1 Homo sapiens 152-156 29796178-4 2018 Induction of HO-1 protein expression was mimicked by SB203580, another p38 MAPK inhibitor, but not by SB202474, an inactive structural analogue of p38 MAPK inhibitors. SB 203580 53-61 heme oxygenase 1 Homo sapiens 13-17 29796178-5 2018 HO-1 induction by both SB202190 and SB203580 was also demonstrated by analysis of mRNA expression. SB 203580 36-44 heme oxygenase 1 Homo sapiens 0-4 29796178-7 2018 Treatment of cells with tin protoporphyrin IX (SnPPIX), a well-characterised HO-1 enzymatic inhibitor, or HO-1 siRNA left SB202190-modulated metabolic activity and autophagy virtually unaltered but caused a significant reversal of the anti-apoptotic action of SB202190. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 122-130 heme oxygenase 1 Homo sapiens 106-110 29796178-8 2018 Conversely, however, HO-1 expression by SB202190 became completely suppressed by the autophagy inhibitor bafilomycin A1. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 40-48 heme oxygenase 1 Homo sapiens 21-25 29796178-8 2018 Conversely, however, HO-1 expression by SB202190 became completely suppressed by the autophagy inhibitor bafilomycin A1. bafilomycin 105-116 heme oxygenase 1 Homo sapiens 21-25 29796178-10 2018 Collectively, this work demonstrates SB202190 to confer upstream induction of autophagy followed by HO-1 induction resulting in potential protective effects against apoptosis. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 37-45 heme oxygenase 1 Homo sapiens 100-104 29796178-11 2018 On the other hand, our data oppose HO-1 to contribute to SB202190-mediated increases in metabolic activity and autophagy, respectively. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 57-65 heme oxygenase 1 Homo sapiens 35-39 29241092-5 2018 In addition, trehalose increased nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in a p62-dependent manner and enhance expression of its downstream antioxidant factors, heme oxygenase-1 (Ho-1) and nicotinamide adenine dinucleotide phosphate quinone dehydrogenase 1 (Nqo1). Trehalose 13-22 heme oxygenase 1 Homo sapiens 197-213 29854096-5 2018 The effects of cafestol on mitogen-activated protein kinases (MAPK), heme oxygenase-1 (HO-1), and sirtuin 1 (Sirt1) signaling pathways were examined using Western blotting and specific inhibitors. cafestol 15-23 heme oxygenase 1 Homo sapiens 69-85 29854096-8 2018 By contrast, cafestol upregulated cyclic-strain-induced HO-1 and Sirt1 expression. cafestol 13-21 heme oxygenase 1 Homo sapiens 56-60 29854096-10 2018 This is the first study to report that cafestol inhibited cyclic-strain-induced inflammatory molecule secretion, possibly through the activation of HO-1 and Sirt1 in endothelial cells. cafestol 39-47 heme oxygenase 1 Homo sapiens 148-152 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. ammonium ferrous sulfate 144-148 heme oxygenase 1 Homo sapiens 0-16 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. ammonium ferrous sulfate 144-148 heme oxygenase 1 Homo sapiens 18-22 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Biliverdine 155-165 heme oxygenase 1 Homo sapiens 0-16 29525432-1 2018 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to produce three anti-oxidant molecules: carbon monoxide (CO), ferrous ion (Fe2+), and biliverdin. Biliverdine 155-165 heme oxygenase 1 Homo sapiens 18-22 29525432-3 2018 In the present study, we identified marliolide as a novel inducer of HO-1 in human normal keratinocyte HaCaT cells. marliolide 36-46 heme oxygenase 1 Homo sapiens 69-73 29525432-4 2018 Mechanism-based studies demonstrated that the induction of HO-1 by marliolide occurred through activation of NRF2/ARE via direct binding of marliolide to KEAP1. marliolide 67-77 heme oxygenase 1 Homo sapiens 59-63 29525432-4 2018 Mechanism-based studies demonstrated that the induction of HO-1 by marliolide occurred through activation of NRF2/ARE via direct binding of marliolide to KEAP1. marliolide 140-150 heme oxygenase 1 Homo sapiens 59-63 29525432-5 2018 Structure-activity relationship revealed chemical moieties of marliolide critical for induction of HO-1, which renders a support for Michael reaction as a potential mechanism of action. marliolide 62-72 heme oxygenase 1 Homo sapiens 99-103 29679467-7 2018 We also showed that nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/heme oxygenase 1 (HO-1) and nuclear factor kappa B (NF-kappaB) signaling was critical for the salidroside-mediated beneficial regulation. rhodioloside 165-176 heme oxygenase 1 Homo sapiens 71-87 29674687-3 2018 In human arterial and venous endothelial cells (EC), and in contrast to ibuprofen and naproxen, celecoxib induced the protective protein heme oxygenase-1 (HO-1). Celecoxib 96-105 heme oxygenase 1 Homo sapiens 137-153 29674687-3 2018 In human arterial and venous endothelial cells (EC), and in contrast to ibuprofen and naproxen, celecoxib induced the protective protein heme oxygenase-1 (HO-1). Celecoxib 96-105 heme oxygenase 1 Homo sapiens 155-159 29674687-4 2018 Celecoxib derivative 2,5-dimethyl-celecoxib (DMC) which lacks COX-2 inhibition also upregulated HO-1, implicating a COX-2-independent mechanism. Celecoxib 0-9 heme oxygenase 1 Homo sapiens 96-100 29674687-4 2018 Celecoxib derivative 2,5-dimethyl-celecoxib (DMC) which lacks COX-2 inhibition also upregulated HO-1, implicating a COX-2-independent mechanism. 2,5-dimethylcelecoxib 21-43 heme oxygenase 1 Homo sapiens 96-100 29674687-4 2018 Celecoxib derivative 2,5-dimethyl-celecoxib (DMC) which lacks COX-2 inhibition also upregulated HO-1, implicating a COX-2-independent mechanism. 2,5-dimethylcelecoxib 45-48 heme oxygenase 1 Homo sapiens 96-100 29674687-9 2018 This attenuated VCAM-1 upregulation via induction of HO-1, a response reproduced by DMC but not ibuprofen or naproxen. 2,5-dimethylcelecoxib 84-87 heme oxygenase 1 Homo sapiens 53-57 29765448-8 2018 Moreover, it was demonstrated that HO-1 overexpression significantly inhibited the hypoxia-promoted STAT3 phosphorylation (Tyr 705). Tyrosine 123-126 heme oxygenase 1 Homo sapiens 35-39 29543451-4 2018 With the exception of DeltaNOD, the presence of an intact catechol structure was obligatory for the inhibition of VCAM-1 and the induction of HO-1 expression. catechol 58-66 heme oxygenase 1 Homo sapiens 142-146 29524413-0 2018 Heme oxygenase-1/carbon monoxide axis suppresses transforming growth factor-beta1-induced growth inhibition by increasing ERK1/2-mediated phosphorylation of Smad3 at Thr-179 in human hepatocellular carcinoma cell lines. Carbon Monoxide 17-32 heme oxygenase 1 Homo sapiens 0-16 29524413-0 2018 Heme oxygenase-1/carbon monoxide axis suppresses transforming growth factor-beta1-induced growth inhibition by increasing ERK1/2-mediated phosphorylation of Smad3 at Thr-179 in human hepatocellular carcinoma cell lines. Threonine 166-169 heme oxygenase 1 Homo sapiens 0-16 29524413-4 2018 Moreover, TGF-beta1-induced cell cycle arrest and up-regulation of cyclin-dependent kinase inhibitors in HCC cell lines were significantly attenuated by overexpression of HO-1 or treatment with tricarbonyldichlororuthenium(II) dimer ([Ru(CO)3Cl2]2, suggesting an inhibitory role of the HO-1/CO axis in TGF-beta signaling to growth inhibition in HCC cell lines. tricarbonyldichlororuthenium(ii 194-225 heme oxygenase 1 Homo sapiens 286-290 29524413-4 2018 Moreover, TGF-beta1-induced cell cycle arrest and up-regulation of cyclin-dependent kinase inhibitors in HCC cell lines were significantly attenuated by overexpression of HO-1 or treatment with tricarbonyldichlororuthenium(II) dimer ([Ru(CO)3Cl2]2, suggesting an inhibitory role of the HO-1/CO axis in TGF-beta signaling to growth inhibition in HCC cell lines. [ru(co)3cl2 234-245 heme oxygenase 1 Homo sapiens 171-175 29524413-6 2018 Additional experiments revealed that HO-1/CO axis selectively induces phosphorylation of Smad3 at Thr-179 residue in the linker region through activation of extracellular signal-activated kinase (ERK) 1/2. Threonine 98-101 heme oxygenase 1 Homo sapiens 37-44 29524413-7 2018 Transfection with a phospho-deficient Smad3 (T179A) mutant or treatment with FR180204, a specific inhibitor for ERK1/2, significantly reversed the inhibitory effects of HO-1 and [Ru(CO)3Cl2]2 on cell cycle arrest induced by TGF-beta1. FR 180204 77-85 heme oxygenase 1 Homo sapiens 169-173 29524413-8 2018 These findings for the first time demonstrate that HO-1/CO axis confer resistance of HCC cells to TGF-beta growth inhibitory signal by increasing Smad3 phosphorylation at Thr-179 via ERK1/2 pathway. Threonine 171-174 heme oxygenase 1 Homo sapiens 51-55 29652826-0 2018 Cardiovascular Mechanisms of Action of Anthocyanins May Be Associated with the Impact of Microbial Metabolites on Heme Oxygenase-1 in Vascular Smooth Muscle Cells. Anthocyanins 39-51 heme oxygenase 1 Homo sapiens 114-130 29652826-2 2018 We aimed to explore the effects of microbial metabolites common to anthocyanins and other flavonoids on vascular smooth muscle heme oxygenase-1 (HO-1) expression. Anthocyanins 67-79 heme oxygenase 1 Homo sapiens 127-143 29652826-2 2018 We aimed to explore the effects of microbial metabolites common to anthocyanins and other flavonoids on vascular smooth muscle heme oxygenase-1 (HO-1) expression. Anthocyanins 67-79 heme oxygenase 1 Homo sapiens 145-149 29652826-2 2018 We aimed to explore the effects of microbial metabolites common to anthocyanins and other flavonoids on vascular smooth muscle heme oxygenase-1 (HO-1) expression. Flavonoids 90-100 heme oxygenase 1 Homo sapiens 127-143 29652826-2 2018 We aimed to explore the effects of microbial metabolites common to anthocyanins and other flavonoids on vascular smooth muscle heme oxygenase-1 (HO-1) expression. Flavonoids 90-100 heme oxygenase 1 Homo sapiens 145-149 29652826-5 2018 The present study demonstrates that phenolic metabolites of anthocyanins differentially affect HO-1 activity, often having additive, synergistic or nullifying effects. Anthocyanins 60-72 heme oxygenase 1 Homo sapiens 95-99 29437594-2 2018 Patrolling monocytes, which normally scavenge damaged cells and debris from the vasculature, express higher levels of anti-inflammatory heme oxygenase 1 (HO-1), a heme degrading enzyme. Heme 136-140 heme oxygenase 1 Homo sapiens 154-158 29437594-4 2018 We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. Heme 202-206 heme oxygenase 1 Homo sapiens 95-99 29437594-4 2018 We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. Heme 202-206 heme oxygenase 1 Homo sapiens 101-107 29437594-4 2018 We found that a mean 37% of patrolling monocytes from SCD patients express very high levels of HO-1 (HO-1hi) vs 6% in healthy controls and demonstrated that HO-1hi expression was dependent on uptake of heme-exposed endothelium. Heme 202-206 heme oxygenase 1 Homo sapiens 157-163 29765448-9 2018 Conclusions: HO-1 was overexpressed in PE placenta, in association with reduced STAT3 phosphorylation (Tyr 705). Tyrosine 103-106 heme oxygenase 1 Homo sapiens 13-17 29765448-11 2018 Thus, we speculate that overexpressed HO-1 might contribute to the reduced STAT3 phosphorylation (Tyr 705) and the pathogenesis of preeclampsia. Tyrosine 98-101 heme oxygenase 1 Homo sapiens 38-42 29471244-0 2018 Two sesquiterpene aminoquinones protect against oxidative injury in HaCaT keratinocytes via activation of AMPKalpha/ERK-Nrf2/ARE/HO-1 signaling. sesquiterpene aminoquinones 4-31 heme oxygenase 1 Homo sapiens 129-133 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Curcumin 0-8 heme oxygenase 1 Homo sapiens 172-188 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Curcumin 0-8 heme oxygenase 1 Homo sapiens 190-194 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Curcumin 0-8 heme oxygenase 1 Homo sapiens 245-249 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Polychlorinated Dibenzodioxins 36-40 heme oxygenase 1 Homo sapiens 172-188 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Polychlorinated Dibenzodioxins 36-40 heme oxygenase 1 Homo sapiens 190-194 29191105-3 2018 Curcumin dose-dependently inhibited TCDD-induced expression of phase I enzyme cytochrome P450 1A1 (CYP1A1) and phase II enzymes NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase 1 (HO-1) but not tert-butyl hydroquinone-induced NQO1 and HO-1, suggesting that curcumin inhibited only AhR pathway, but not Nrf2 one directly. Polychlorinated Dibenzodioxins 36-40 heme oxygenase 1 Homo sapiens 245-249 29471244-11 2018 SIGNIFICANCE: DAs provided cytoprotective effects against H2O2-induced cytotoxicity by activation of the Nrf2/ARE/HO-1 pathway via phosphorylation of AMPKalpha and ERK. Hydrogen Peroxide 58-62 heme oxygenase 1 Homo sapiens 114-118 29421327-1 2018 Our previous study has shown heme oxygenase-1 (HO-1) protects human lens epithelial cells (LECs) against H2O2-induced oxidative stress and apoptosis. Hydrogen Peroxide 105-109 heme oxygenase 1 Homo sapiens 29-45 29351417-9 2018 Zn2+ also regulated inflammation-related key molecules such as heme oxygenase-1, selectin L, IL-10, and platelet endothelial cell adhesion molecule 1, as well as vascular tone-related prostaglandin I2 synthase and nitric oxide synthase-3. Zinc 0-4 heme oxygenase 1 Homo sapiens 63-79 29339440-4 2018 Single-cell RNA sequencing analysis, tumor microarray, and clinical survival data from breast cancer patients were used to support the clinical relevance of our observations.Results: We demonstrate that SnMP inhibits immune suppression of chemotherapy-elicited CD8+ T cells by targeting myeloid HO-1 activity in the tumor microenvironment. tin mesoporphyrin 203-207 heme oxygenase 1 Homo sapiens 295-299 29421327-1 2018 Our previous study has shown heme oxygenase-1 (HO-1) protects human lens epithelial cells (LECs) against H2O2-induced oxidative stress and apoptosis. Hydrogen Peroxide 105-109 heme oxygenase 1 Homo sapiens 47-51 29436680-16 2018 Overall, these data indicated that curcumin activates the Nrf2-ARE signaling pathway and upregulates HO-1 expression, which mediates SR-BI and ABCA1 expression and thereby increases cholesterol efflux. Cholesterol 182-193 heme oxygenase 1 Homo sapiens 101-105 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Bilirubin 108-117 heme oxygenase 1 Homo sapiens 0-16 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Carbon Monoxide 119-134 heme oxygenase 1 Homo sapiens 0-16 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Carbon Monoxide 136-138 heme oxygenase 1 Homo sapiens 0-16 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Iron 149-153 heme oxygenase 1 Homo sapiens 0-16 29302043-3 2018 Heme oxygenase-1 (HO) is the rate limiting enzyme in heme metabolism leading to the equimolar production of bilirubin, carbon monoxide (CO) and free iron (Fe). Iron 155-157 heme oxygenase 1 Homo sapiens 0-16 29436680-0 2018 Curcumin increases cholesterol efflux via heme oxygenase-1-mediated ABCA1 and SR-BI expression in macrophages. Curcumin 0-8 heme oxygenase 1 Homo sapiens 42-58 29436680-0 2018 Curcumin increases cholesterol efflux via heme oxygenase-1-mediated ABCA1 and SR-BI expression in macrophages. Cholesterol 19-30 heme oxygenase 1 Homo sapiens 42-58 29436680-9 2018 Additionally, curcumin significantly upregulated HO-1 expression. Curcumin 14-22 heme oxygenase 1 Homo sapiens 49-53 29436680-10 2018 The HO-1 inhibitor (zinc protoporphyrin) partly blocked this effect. zinc protoporphyrin 20-39 heme oxygenase 1 Homo sapiens 4-8 29436680-12 2018 HO-1 small interfering (si)RNA partly abolished the increased SR-BI and ABCA1 expression induced by curcumin. Curcumin 100-108 heme oxygenase 1 Homo sapiens 0-4 29436680-14 2018 Nrf2 siRNA successfully inhibited the curcumin-induced HO-1 expression. Curcumin 38-46 heme oxygenase 1 Homo sapiens 55-59 29436680-16 2018 Overall, these data indicated that curcumin activates the Nrf2-ARE signaling pathway and upregulates HO-1 expression, which mediates SR-BI and ABCA1 expression and thereby increases cholesterol efflux. Curcumin 35-43 heme oxygenase 1 Homo sapiens 101-105 29411263-0 2018 Naringenin Exerts Anti-inflammatory Effects in Paraquat-Treated SH-SY5Y Cells Through a Mechanism Associated with the Nrf2/HO-1 Axis. naringenin 0-10 heme oxygenase 1 Homo sapiens 123-127 28456938-6 2018 Additionally, resveratrol prevented the LPS-induced increase in the intracellular reactive oxygen species (ROS) as well as the decrease in glutathione (GSH) levels and in glutamate cysteine ligase (GCL) activity, through Nrf2/HO-1 signaling pathways. Resveratrol 14-25 heme oxygenase 1 Homo sapiens 226-230 28456938-4 2018 In this sense, we have demonstrated that resveratrol has a protective effect on oligodendroglial functionality against lipopolysaccharide (LPS)-mediated cytotoxicity and that its glioprotective mechanism involves the nuclear factor erythroid 2-related factor 2 (Nrf2) and HO-1 pathways. Resveratrol 41-52 heme oxygenase 1 Homo sapiens 272-276 28497200-7 2018 Furthermore, BTZ significantly induced heme oxygenase-1 as a result of increased oxidative stress and such overexpression was prevented by concomitant treatment with ALA. Bortezomib 13-16 heme oxygenase 1 Homo sapiens 39-55 28497200-7 2018 Furthermore, BTZ significantly induced heme oxygenase-1 as a result of increased oxidative stress and such overexpression was prevented by concomitant treatment with ALA. ala 166-169 heme oxygenase 1 Homo sapiens 39-55 29411263-0 2018 Naringenin Exerts Anti-inflammatory Effects in Paraquat-Treated SH-SY5Y Cells Through a Mechanism Associated with the Nrf2/HO-1 Axis. Paraquat 47-55 heme oxygenase 1 Homo sapiens 123-127 29411263-2 2018 NGN is a potent inducer of the nuclear factor erythroid 2-related factor 2 (Nrf2) and upregulates the expression of heme oxygenase-1 (HO-1), an enzyme exhibiting both antioxidant and anti-inflammatory effects. naringenin 0-3 heme oxygenase 1 Homo sapiens 116-132 29411263-2 2018 NGN is a potent inducer of the nuclear factor erythroid 2-related factor 2 (Nrf2) and upregulates the expression of heme oxygenase-1 (HO-1), an enzyme exhibiting both antioxidant and anti-inflammatory effects. naringenin 0-3 heme oxygenase 1 Homo sapiens 134-138 29411263-9 2018 The anti-apoptotic and anti-inflammatory effects promoted by NGN were abolished by ZnPP IX (a specific inhibitor of HO-1) or by knockdown of Nrf2 by small interfering RNA (siRNA). naringenin 61-64 heme oxygenase 1 Homo sapiens 116-120 29411263-9 2018 The anti-apoptotic and anti-inflammatory effects promoted by NGN were abolished by ZnPP IX (a specific inhibitor of HO-1) or by knockdown of Nrf2 by small interfering RNA (siRNA). zinc protoporphyrin 83-90 heme oxygenase 1 Homo sapiens 116-120 29411263-10 2018 Therefore, NGN induced anti-inflammatory effects in PQ-treated SH-SY5Y cells by a mechanism associated with the Nrf2/HO-1 signaling pathway. naringenin 11-14 heme oxygenase 1 Homo sapiens 117-121 29526395-0 2018 Up-regulation of HO-1 by Nrf2 activation protects against palmitic acid-induced ROS increase in human neuroblastoma BE(2)-M17 cells. Palmitic Acid 58-71 heme oxygenase 1 Homo sapiens 17-21 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Reactive Oxygen Species 110-113 heme oxygenase 1 Homo sapiens 53-57 29411263-10 2018 Therefore, NGN induced anti-inflammatory effects in PQ-treated SH-SY5Y cells by a mechanism associated with the Nrf2/HO-1 signaling pathway. Paraquat 52-54 heme oxygenase 1 Homo sapiens 117-121 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Reactive Oxygen Species 205-208 heme oxygenase 1 Homo sapiens 53-57 29526395-0 2018 Up-regulation of HO-1 by Nrf2 activation protects against palmitic acid-induced ROS increase in human neuroblastoma BE(2)-M17 cells. Beryllium 116-118 heme oxygenase 1 Homo sapiens 17-21 29743974-0 2018 Simvastatin Treatment Upregulates HO-1 in Patients with Abdominal Aortic Aneurysm but Independently of Nrf2. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 34-38 29526395-2 2018 Extracellular signal regulated kinase (ERK)/nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) is a ROS response pathway. Reactive Oxygen Species 124-127 heme oxygenase 1 Homo sapiens 95-111 29526395-2 2018 Extracellular signal regulated kinase (ERK)/nuclear factor erythroid-2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) is a ROS response pathway. Reactive Oxygen Species 124-127 heme oxygenase 1 Homo sapiens 113-117 29526395-4 2018 We hypothesized that the up-regulation of HO-1 is compensatory for response to fatty acid-induced oxidative stress but not enough to reduce ROS levels. Fatty Acids 79-89 heme oxygenase 1 Homo sapiens 42-46 29526395-5 2018 We evaluated the anti-ROS effect of HO-1 and the involved pathway in palmitic acid (PA)-treated human neuroblastoma BE(2)-M17 cells. Reactive Oxygen Species 22-25 heme oxygenase 1 Homo sapiens 36-40 29526395-5 2018 We evaluated the anti-ROS effect of HO-1 and the involved pathway in palmitic acid (PA)-treated human neuroblastoma BE(2)-M17 cells. Palmitic Acid 84-86 heme oxygenase 1 Homo sapiens 36-40 29526395-5 2018 We evaluated the anti-ROS effect of HO-1 and the involved pathway in palmitic acid (PA)-treated human neuroblastoma BE(2)-M17 cells. Beryllium 116-118 heme oxygenase 1 Homo sapiens 36-40 29526395-6 2018 As expected, PA-induced ROS increase was accompanied by activation of the ERK-Nrf2-HO-1 pathway, as demonstrated by an increase in ERK phosphorylation, Nrf2 phosphorylation and nuclear accumulation, and HO-1 expression at the mRNA and protein levels, in a PA-dose-dependent manner. Palmitic Acid 13-15 heme oxygenase 1 Homo sapiens 83-87 29526395-6 2018 As expected, PA-induced ROS increase was accompanied by activation of the ERK-Nrf2-HO-1 pathway, as demonstrated by an increase in ERK phosphorylation, Nrf2 phosphorylation and nuclear accumulation, and HO-1 expression at the mRNA and protein levels, in a PA-dose-dependent manner. Palmitic Acid 13-15 heme oxygenase 1 Homo sapiens 203-207 29526395-6 2018 As expected, PA-induced ROS increase was accompanied by activation of the ERK-Nrf2-HO-1 pathway, as demonstrated by an increase in ERK phosphorylation, Nrf2 phosphorylation and nuclear accumulation, and HO-1 expression at the mRNA and protein levels, in a PA-dose-dependent manner. Palmitic Acid 256-258 heme oxygenase 1 Homo sapiens 83-87 29526395-7 2018 In contrast, administration of the ROS scavenger NAC significantly reduced the levels of PA-regulated ROS and HO-1 protein. Reactive Oxygen Species 35-38 heme oxygenase 1 Homo sapiens 110-114 29526395-7 2018 In contrast, administration of the ROS scavenger NAC significantly reduced the levels of PA-regulated ROS and HO-1 protein. Palmitic Acid 89-91 heme oxygenase 1 Homo sapiens 110-114 29526395-8 2018 However, the ERK inhibitor U0126 not only reversed the activating effect of PA on the ERK-Nrf2-HO-1 pathway but also aggravated PA-induced ROS. U 0126 27-32 heme oxygenase 1 Homo sapiens 95-99 29526395-8 2018 However, the ERK inhibitor U0126 not only reversed the activating effect of PA on the ERK-Nrf2-HO-1 pathway but also aggravated PA-induced ROS. Palmitic Acid 76-78 heme oxygenase 1 Homo sapiens 95-99 29526395-9 2018 Furthermore, the Nrf2-specific activator NK-252 significantly increased PA-up-regulated HO-1 protein and alleviated PA-induced ROS. Palmitic Acid 72-74 heme oxygenase 1 Homo sapiens 88-92 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Palmitic Acid 61-63 heme oxygenase 1 Homo sapiens 53-57 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Reactive Oxygen Species 205-208 heme oxygenase 1 Homo sapiens 143-147 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Palmitic Acid 223-225 heme oxygenase 1 Homo sapiens 53-57 29526395-10 2018 Therefore, our results suggest that up-regulation of HO-1 in PA-treated neurons is a compensatory response to ROS increase and that increasing HO-1 expression by Nrf2 activation can prevent the process of ROS production in PA-treated neurons. Palmitic Acid 223-225 heme oxygenase 1 Homo sapiens 143-147 29425821-0 2018 MMP9 is involved in HO-1-mediated upregulation of apical junctional complex in Caco-2 cells under oxygen-glucose deprivation. oxygen-glucose 98-112 heme oxygenase 1 Homo sapiens 20-24 29425821-2 2018 This study was the first to explore the expression of apical junctional complex (AJC) induced by heme oxygenase-1 (HO-1) in Caco-2 cells in oxygen-glucose deprivation (OGD) models. oxygen-glucose 140-154 heme oxygenase 1 Homo sapiens 97-113 29425821-2 2018 This study was the first to explore the expression of apical junctional complex (AJC) induced by heme oxygenase-1 (HO-1) in Caco-2 cells in oxygen-glucose deprivation (OGD) models. oxygen-glucose 140-154 heme oxygenase 1 Homo sapiens 115-119 29454190-0 2018 Potholing of the hydrophobic heme oxygenase-1 western region for the search of potent and selective imidazole-based inhibitors. imidazole 100-109 heme oxygenase 1 Homo sapiens 29-45 29743974-6 2018 Simvastatin treatment increased HO-1 protein level in AAA, both in endothelial cells (ECs) and in smooth muscle cells (SMCs), but increased Nrf2 localization was restricted only to vasa vasorum. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 32-36 29743974-8 2018 In vitro studies showed that simvastatin raises HO-1 protein level slightly in ECs and to much higher extent in SMCs, which is not related to Nrf2/ARE activation, although HMOX1 expression is upregulated by simvastatin in both cell types. Simvastatin 29-40 heme oxygenase 1 Homo sapiens 48-52 29743974-8 2018 In vitro studies showed that simvastatin raises HO-1 protein level slightly in ECs and to much higher extent in SMCs, which is not related to Nrf2/ARE activation, although HMOX1 expression is upregulated by simvastatin in both cell types. Simvastatin 207-218 heme oxygenase 1 Homo sapiens 172-177 29743974-9 2018 In conclusion, simvastatin-induced modulation of HO-1 level in ECs and SMCs in vitro is not related to Nrf2/ARE activity. Simvastatin 15-26 heme oxygenase 1 Homo sapiens 49-53 29355553-6 2018 Reb-A treatment induced nuclear factor erythroid-derived 2-like 2 (Nrf2) activation and antioxidant response element activity, as well as the expression of heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). rebaudioside A 0-5 heme oxygenase 1 Homo sapiens 156-172 29619425-1 2018 Background: Heme oxygenase-1 (HO-1) is a protein secreted by immune cells as a part of immune response mechanism.HO-1 can be induced by variety agents that causingoxidative stress, such as exposure to 100% oxygenat2,4 ATA pressure.It plays a vital role in maintaining cellular homeostasis.This study was conducted to identify the effect of hyperbaric oxygen exposure in cultured ofPBMCthat infected by HIV-1. Oxygen 17-23 heme oxygenase 1 Homo sapiens 30-34 29619425-1 2018 Background: Heme oxygenase-1 (HO-1) is a protein secreted by immune cells as a part of immune response mechanism.HO-1 can be induced by variety agents that causingoxidative stress, such as exposure to 100% oxygenat2,4 ATA pressure.It plays a vital role in maintaining cellular homeostasis.This study was conducted to identify the effect of hyperbaric oxygen exposure in cultured ofPBMCthat infected by HIV-1. Oxygen 17-23 heme oxygenase 1 Homo sapiens 113-117 29274359-8 2018 The ferroptotic process induced by hHO-1 overexpression further indicated that HO-1 is a key mediator of BAY-induced ferroptosis that operates through cellular redox regulation and iron accumulation. Iron 181-185 heme oxygenase 1 Homo sapiens 35-40 29165873-8 2018 Concomitantly, the HO-1 byproduct CO and the cGMP analogue 8-bromoguanosine 3",5"-cyclic monophosphate (8-Br-cGMP) increased MKP-5 expression, and pretreatment with CO and 8-Br-cGMP inhibited hypoxia-induced HIF-1alpha and ET-1 expression. 8-bromoguanosino-3',5'-cyclic monophosphorothioate 104-113 heme oxygenase 1 Homo sapiens 19-23 29165873-0 2018 Andrographolide inhibits hypoxia-induced hypoxia-inducible factor 1alpha and endothelin 1 expression through the heme oxygenase 1/CO/cGMP/MKP-5 pathways in EA.hy926 cells. andrographolide 0-15 heme oxygenase 1 Homo sapiens 113-129 29165873-8 2018 Concomitantly, the HO-1 byproduct CO and the cGMP analogue 8-bromoguanosine 3",5"-cyclic monophosphate (8-Br-cGMP) increased MKP-5 expression, and pretreatment with CO and 8-Br-cGMP inhibited hypoxia-induced HIF-1alpha and ET-1 expression. Carbon Monoxide 34-36 heme oxygenase 1 Homo sapiens 19-23 29165873-7 2018 Andrographolide enhanced HO-1 and MKP-5 expression and cellular cGMP content in addition to inhibiting hypoxia-induced ROS generation. andrographolide 0-15 heme oxygenase 1 Homo sapiens 25-29 29165873-8 2018 Concomitantly, the HO-1 byproduct CO and the cGMP analogue 8-bromoguanosine 3",5"-cyclic monophosphate (8-Br-cGMP) increased MKP-5 expression, and pretreatment with CO and 8-Br-cGMP inhibited hypoxia-induced HIF-1alpha and ET-1 expression. 8-bromoguanosino-3',5'-cyclic monophosphorothioate 172-181 heme oxygenase 1 Homo sapiens 19-23 29165873-9 2018 Transfection of HO-1 siRNA or pretreatment with the HO-1 inhibitor ZnPP-9 or 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, a specific inhibitor of soluble guanylate cyclase, reduced andrographolide-induced MKP-5 expression. zinc protoporphyrin 67-73 heme oxygenase 1 Homo sapiens 52-56 29165873-11 2018 The inhibition of hypoxia-induced HIF-1alpha and ET-1 expression by andrographolide is likely associated with HO-1/CO/cGMP/MKP-5 pathways, which is involved in inhibiting hypoxia-induced p38 MAPK activation. andrographolide 68-83 heme oxygenase 1 Homo sapiens 110-117 29165873-11 2018 The inhibition of hypoxia-induced HIF-1alpha and ET-1 expression by andrographolide is likely associated with HO-1/CO/cGMP/MKP-5 pathways, which is involved in inhibiting hypoxia-induced p38 MAPK activation. Cyclic GMP 118-122 heme oxygenase 1 Homo sapiens 110-117 29317217-0 2018 Crucial role of HO-1/IRF4-dependent apoptosis induced by panobinostat and lenalidomide in multiple myeloma. Panobinostat 57-69 heme oxygenase 1 Homo sapiens 16-20 29168081-0 2018 High Glucose Stimulates Expression of MFHAS1 to Mitigate Inflammation via Akt/HO-1 Pathway in Human Umbilical Vein Endothelial Cells. Glucose 5-12 heme oxygenase 1 Homo sapiens 78-82 29168081-6 2018 Our results indicated that MFHAS1 deadened high-glucose induced inflammation by activating AKT/HO-1 pathway, suggesting that MFHAS1 may act as a new therapeutic target of diabetes mellitus. high-glucose 43-55 heme oxygenase 1 Homo sapiens 95-99 29470442-9 2018 Induction of HO-1 induced by Pc was suppressed by Go6976, a selective inhibitor of PKC alpha/beta II. Go 6976 50-56 heme oxygenase 1 Homo sapiens 13-17 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 heme oxygenase 1 Homo sapiens 170-174 29317217-10 2018 LBH589 and lenalidomide exerted synergistic effects, and LBH589 reversed the efficacy of lenalidomide on the resistance of CD138+ primary MM cells, in part due to simultaneous suppression of HO-1, IRF4 and MYC. Panobinostat 57-63 heme oxygenase 1 Homo sapiens 191-195 29317217-10 2018 LBH589 and lenalidomide exerted synergistic effects, and LBH589 reversed the efficacy of lenalidomide on the resistance of CD138+ primary MM cells, in part due to simultaneous suppression of HO-1, IRF4 and MYC. Lenalidomide 89-101 heme oxygenase 1 Homo sapiens 191-195 29094331-0 2018 Pterostilbene protects against uraemia serum-induced endothelial cell damage via activation of Keap1/Nrf2/HO-1 signaling. pterostilbene 0-13 heme oxygenase 1 Homo sapiens 106-110 29094331-9 2018 Collectively, our study demonstrated that PT is effective in reducing US-evoked endothelial cell dysfunction via suppression of oxidative/nitrative stress and inflammatory response, which at least partly depended on Keap1/Nrf2/HO-1 signaling pathway. pterostilbene 42-44 heme oxygenase 1 Homo sapiens 227-231 28861811-7 2018 Furthermore, neurotoxicity of lopinavir was blocked by pharmacological augmentation of the endogenous antioxidant heme oxygenase-1 (HO-1), expanding our previous finding that protease inhibitor-induced neurotoxicity was mediated by oxidative stress. Lopinavir 30-39 heme oxygenase 1 Homo sapiens 114-130 29278300-6 2018 Pretreatment with the phenolic acids for 18 h results in a significant upregulation of the Nrf2-regulated antioxidant response proteins heme oxygenase 1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), following 6 h exposure to 2.5 mum H2 O2 . phenolic acid 22-36 heme oxygenase 1 Homo sapiens 136-152 29278300-6 2018 Pretreatment with the phenolic acids for 18 h results in a significant upregulation of the Nrf2-regulated antioxidant response proteins heme oxygenase 1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), following 6 h exposure to 2.5 mum H2 O2 . phenolic acid 22-36 heme oxygenase 1 Homo sapiens 154-158 29278300-6 2018 Pretreatment with the phenolic acids for 18 h results in a significant upregulation of the Nrf2-regulated antioxidant response proteins heme oxygenase 1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), following 6 h exposure to 2.5 mum H2 O2 . Hydrogen Peroxide 249-254 heme oxygenase 1 Homo sapiens 136-152 29278300-6 2018 Pretreatment with the phenolic acids for 18 h results in a significant upregulation of the Nrf2-regulated antioxidant response proteins heme oxygenase 1 (HO-1) and glutamate-cysteine ligase modifier subunit (GCLM), following 6 h exposure to 2.5 mum H2 O2 . Hydrogen Peroxide 249-254 heme oxygenase 1 Homo sapiens 154-158 29307774-7 2018 5-aminolevulinate synthase 1 (ALAS1), Ferrochelatase (FECH), hemeoxygenase 1 (HO-1) and ATP binding cassette subfamily G member 2 (ABCG2)) which affect production of PpIX was assessed. protoporphyrin IX 166-170 heme oxygenase 1 Homo sapiens 61-82 29317217-0 2018 Crucial role of HO-1/IRF4-dependent apoptosis induced by panobinostat and lenalidomide in multiple myeloma. Lenalidomide 74-86 heme oxygenase 1 Homo sapiens 16-20 29317217-7 2018 Herein, panobinostat induced acetylation of histone H3K9 and activation of caspase-3 in MM cells, being inversely correlated with the reduction of HO-1/IRF4/MYC protein levels. Panobinostat 8-20 heme oxygenase 1 Homo sapiens 147-151 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. benzoylcarbonyl-aspartyl-glutamyl-valyl-aspartyl-fluoromethyl ketone 7-17 heme oxygenase 1 Homo sapiens 56-60 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. benzoylcarbonyl-aspartyl-glutamyl-valyl-aspartyl-fluoromethyl ketone 7-17 heme oxygenase 1 Homo sapiens 170-174 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Panobinostat 79-91 heme oxygenase 1 Homo sapiens 56-60 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Panobinostat 79-91 heme oxygenase 1 Homo sapiens 170-174 29317217-8 2018 Adding Z-DEVD-FMK, a caspase-3 inhibitor, abolished the HO-1/IRF4 reduction by panobinostat alone or in combination with lenalidomide, suggesting that caspase-3-mediated HO-1/IRF4/MYC degradation occurred. Lenalidomide 121-133 heme oxygenase 1 Homo sapiens 56-60 28683566-6 2018 Overexpression of miR-92a suppresses expression of heme oxygenase-1 (HO-1), a critical cytoprotective enzyme, whereas inhibition of miR-92a increases HO-1 expression in human umbilical vein ECs (HUVECs) and db/db mouse aortas. mir-92a 18-25 heme oxygenase 1 Homo sapiens 69-73 29381356-0 2018 Astaxanthin Induces the Nrf2/HO-1 Antioxidant Pathway in Human Umbilical Vein Endothelial Cells by Generating Trace Amounts of ROS. astaxanthine 0-11 heme oxygenase 1 Homo sapiens 29-33 29381356-5 2018 In addition, astaxanthin increased the mRNA levels of phase II enzymes HO-1 and also promoted GSH-Px enzyme activity. astaxanthine 13-24 heme oxygenase 1 Homo sapiens 71-75 29381356-7 2018 Knockdown of Nrf-2 by small interfering RNA inhibited HO-1 mRNA expression by 60%, indicating that the Nrf-2/ARE signaling pathway is activated by astaxanthin. astaxanthine 147-158 heme oxygenase 1 Homo sapiens 54-58 29381356-8 2018 Our results suggest that astaxanthin activates the Nrf-2/HO-1 antioxidant pathway by generating small amounts of ROS. astaxanthine 25-36 heme oxygenase 1 Homo sapiens 57-61 29381356-8 2018 Our results suggest that astaxanthin activates the Nrf-2/HO-1 antioxidant pathway by generating small amounts of ROS. Reactive Oxygen Species 113-116 heme oxygenase 1 Homo sapiens 57-61 28683566-6 2018 Overexpression of miR-92a suppresses expression of heme oxygenase-1 (HO-1), a critical cytoprotective enzyme, whereas inhibition of miR-92a increases HO-1 expression in human umbilical vein ECs (HUVECs) and db/db mouse aortas. mir-92a 18-25 heme oxygenase 1 Homo sapiens 51-67 29541348-0 2018 Protective Effect of Boswellic Acids against Doxorubicin-Induced Hepatotoxicity: Impact on Nrf2/HO-1 Defense Pathway. boswellic acid 21-36 heme oxygenase 1 Homo sapiens 96-100 29643980-5 2018 In human keratinocytes, we found that perillaldehyde (1) inhibited BaP-induced AHR activation and ROS production, (2) inhibited BaP/AHR-mediated release of the CCL2 chemokine, and (3) activated the NRF2/HO1 antioxidant pathway. perillaldehyde 38-52 heme oxygenase 1 Homo sapiens 203-206 29541348-0 2018 Protective Effect of Boswellic Acids against Doxorubicin-Induced Hepatotoxicity: Impact on Nrf2/HO-1 Defense Pathway. Doxorubicin 45-56 heme oxygenase 1 Homo sapiens 96-100 29541348-8 2018 BAs increased the Nrf2 and HO-1 expression, which provided protection against DOX-induced oxidative insult. Doxorubicin 78-81 heme oxygenase 1 Homo sapiens 27-31 29394034-7 2018 Furthermore, Fe-binding and Fe-regulatory proteins, such as hepcidin, lipocalin-2/NGAL, heme oxygenase-1, ferritin, and iron-sulfur clusters can display antitumor properties under specific conditions and in particular cancer types. Iron 13-15 heme oxygenase 1 Homo sapiens 88-104 29560114-9 2018 Intriguingly, we observed that treatment with anti-tumoral epigenetic drugs like LBH-589 (Panobinostat) and Curcumin induced the expression of linc-PINT and HMOX1 in ALL. Curcumin 108-116 heme oxygenase 1 Homo sapiens 157-162 29246853-7 2018 Further experiments demonstrated that cafestol increased translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) and expression of enhanced heme oxygenase-1. cafestol 38-46 heme oxygenase 1 Homo sapiens 152-168 29246853-9 2018 Cafestol"s inhibitory effects on interleukin-8 expression and cellular proliferation induced by urotensin II were significantly abrogated by heme oxygenase-1 silencing, suggesting it may be involved in mediating the effects of cafestol. cafestol 0-8 heme oxygenase 1 Homo sapiens 141-157 29246853-9 2018 Cafestol"s inhibitory effects on interleukin-8 expression and cellular proliferation induced by urotensin II were significantly abrogated by heme oxygenase-1 silencing, suggesting it may be involved in mediating the effects of cafestol. cafestol 227-235 heme oxygenase 1 Homo sapiens 141-157 29246853-10 2018 This study reports that cafestol inhibits urotensin II-induced interleukin-8 expression and cell proliferation via Nrf2/heme oxygenase-1-dependent mechanism in endothelial cells. cafestol 24-32 heme oxygenase 1 Homo sapiens 120-136 29309760-9 2018 These results suggested that rosiglitazone-induced HO-1 expression is mediated through PKCalpha/AMPKalpha/p38 MAPKalpha/SIRT1-dependent deacetylation of Ac-PGC1alpha and fragmentation of NCoR/PPARgamma activation in HPAEpiCs. Rosiglitazone 29-42 heme oxygenase 1 Homo sapiens 51-55 29309760-0 2018 Heme oxygenase-1 induction by rosiglitazone via PKCalpha/AMPKalpha/p38 MAPKalpha/SIRT1/PPARgamma pathway suppresses lipopolysaccharide-mediated pulmonary inflammation. Rosiglitazone 30-43 heme oxygenase 1 Homo sapiens 0-16 28990726-7 2018 Moreover, DMF was able to induce an activation of manganese superoxide dismutase (MnSOD) and heme-oxygenase-1 (HO-1), decreasing the severity of oxidative stress. Dimethyl Fumarate 10-13 heme oxygenase 1 Homo sapiens 93-109 29309760-1 2018 HO-1 (heme oxygenase-1), an antioxidant enzyme, induced by rosiglitazone (PPAR ligands) can be a potential treatment of inflammation. Rosiglitazone 59-72 heme oxygenase 1 Homo sapiens 0-4 29309760-1 2018 HO-1 (heme oxygenase-1), an antioxidant enzyme, induced by rosiglitazone (PPAR ligands) can be a potential treatment of inflammation. Rosiglitazone 59-72 heme oxygenase 1 Homo sapiens 6-22 29309760-2 2018 However, the mechanisms of rosiglitazone-induced HO-1 expression in human pulmonary alveolar epithelial cells (HPAEpiCs) remain largely unknown. Rosiglitazone 27-40 heme oxygenase 1 Homo sapiens 49-53 29309760-3 2018 In this study, we found that upregulation of HO-1 in vitro or in vivo by rosiglitazone attenuated VCAM-1 gene expression and monocyte adhesion to HPAEpiCs challenged with lipopolysaccharide (LPS). Rosiglitazone 73-86 heme oxygenase 1 Homo sapiens 45-49 29309760-4 2018 The inhibitory effects of rosiglitazone on LPS-mediated responses were reversed by transfection with HO-1 siRNA. Rosiglitazone 26-39 heme oxygenase 1 Homo sapiens 101-105 28681934-3 2018 When human SH-SY5Y neuroblastoma cells were treated with DBL or CAPE, the expression of endoplasmic reticulum (ER) stress-related genes such as HSPA5, HYOU1, DDIT3, and SEC61b increased to a larger extent in response to CAPE treatment, while that of antioxidant genes such as HMOX1, GCLM, and NQO1 increased to a larger extent in response to DBL treatment. 3,4-dihydroxybenzalacetone 57-60 heme oxygenase 1 Homo sapiens 276-281 28681934-3 2018 When human SH-SY5Y neuroblastoma cells were treated with DBL or CAPE, the expression of endoplasmic reticulum (ER) stress-related genes such as HSPA5, HYOU1, DDIT3, and SEC61b increased to a larger extent in response to CAPE treatment, while that of antioxidant genes such as HMOX1, GCLM, and NQO1 increased to a larger extent in response to DBL treatment. caffeic acid phenethyl ester 64-68 heme oxygenase 1 Homo sapiens 276-281 28084593-11 2018 The PB-induced antioxidant and anti-inflammatory effects are dependent on the heme oxygenate-1 (HO-1) enzyme and on the activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), since HO-1 inhibition (with 0.5 muM ZnPP IX) or Nrf2 silencing (with small interfering RNA (siRNA)) abolished the effects of PB. pinocembrin 4-6 heme oxygenase 1 Homo sapiens 217-221 28084593-12 2018 Overall, PB afforded cytoprotection by the Nrf2/HO-1 axis in H2O2-treated SH-SY5Y cells. pinocembrin 9-11 heme oxygenase 1 Homo sapiens 48-52 28084593-0 2018 Pinocembrin Suppresses H2O2-Induced Mitochondrial Dysfunction by a Mechanism Dependent on the Nrf2/HO-1 Axis in SH-SY5Y Cells. pinocembrin 0-11 heme oxygenase 1 Homo sapiens 99-103 28084593-11 2018 The PB-induced antioxidant and anti-inflammatory effects are dependent on the heme oxygenate-1 (HO-1) enzyme and on the activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), since HO-1 inhibition (with 0.5 muM ZnPP IX) or Nrf2 silencing (with small interfering RNA (siRNA)) abolished the effects of PB. pinocembrin 4-6 heme oxygenase 1 Homo sapiens 78-94 28084593-12 2018 Overall, PB afforded cytoprotection by the Nrf2/HO-1 axis in H2O2-treated SH-SY5Y cells. Hydrogen Peroxide 61-65 heme oxygenase 1 Homo sapiens 48-52 28084593-11 2018 The PB-induced antioxidant and anti-inflammatory effects are dependent on the heme oxygenate-1 (HO-1) enzyme and on the activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), since HO-1 inhibition (with 0.5 muM ZnPP IX) or Nrf2 silencing (with small interfering RNA (siRNA)) abolished the effects of PB. pinocembrin 4-6 heme oxygenase 1 Homo sapiens 96-100 29134401-11 2018 Duloxetine also induced nuclear translocation of the Nrf2 and the expression of its target gene, HO-1. Duloxetine Hydrochloride 0-10 heme oxygenase 1 Homo sapiens 97-101 29063347-8 2018 Furthermore, administration of the ERK inhibitor U0126 or transient (siRNA) silencing of Nrf2 blocked the protective effects of taurine on cell viability and expression levels of Nrf2 and heme oxygenase-1 (HO-1) in the CORT model of neuronal damage. U 0126 49-54 heme oxygenase 1 Homo sapiens 188-204 29063347-8 2018 Furthermore, administration of the ERK inhibitor U0126 or transient (siRNA) silencing of Nrf2 blocked the protective effects of taurine on cell viability and expression levels of Nrf2 and heme oxygenase-1 (HO-1) in the CORT model of neuronal damage. Taurine 128-135 heme oxygenase 1 Homo sapiens 188-204 29134401-12 2018 Finally, the HO-1 inhibitor, ZnPP, suppressed the duloxetine protective effect. zinc protoporphyrin 29-33 heme oxygenase 1 Homo sapiens 13-17 29496161-9 2018 RESULTS: Three of the nine compounds predicted as Nrf2 activators by molecular docking, gallic acid (GA), Z-liguistilide (LIG), and senkyunolide A (SA), were confirmed with highest potency of increasing the Nrf2/ARE promoter activity and upregulating the expression of Hmox1, Nqo1 and Slc7a11. Gallic Acid 88-99 heme oxygenase 1 Homo sapiens 269-274 29134401-0 2018 Duloxetine Protects Human Neuroblastoma Cells from Oxidative Stress-Induced Cell Death Through Akt/Nrf-2/HO-1 Pathway. Duloxetine Hydrochloride 0-10 heme oxygenase 1 Homo sapiens 105-109 29134401-5 2018 The phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) inhibitor LY294002 (10 microM) and the heme oxygenase 1 (HO-1) inhibitor zinc protoporphyrin IX-ZnPP (5 microM) were added to cultures 1 h prior duloxetine treatments. zinc protoporphyrin ix-znpp 135-162 heme oxygenase 1 Homo sapiens 119-123 29496161-9 2018 RESULTS: Three of the nine compounds predicted as Nrf2 activators by molecular docking, gallic acid (GA), Z-liguistilide (LIG), and senkyunolide A (SA), were confirmed with highest potency of increasing the Nrf2/ARE promoter activity and upregulating the expression of Hmox1, Nqo1 and Slc7a11. 3-N-butyl-4,5-dihydrophthalide 132-146 heme oxygenase 1 Homo sapiens 269-274 29496161-9 2018 RESULTS: Three of the nine compounds predicted as Nrf2 activators by molecular docking, gallic acid (GA), Z-liguistilide (LIG), and senkyunolide A (SA), were confirmed with highest potency of increasing the Nrf2/ARE promoter activity and upregulating the expression of Hmox1, Nqo1 and Slc7a11. 3-N-butyl-4,5-dihydrophthalide 148-150 heme oxygenase 1 Homo sapiens 269-274 29134401-8 2018 Furthermore, the Nrf2 and HO-1 mRNA expression was carried out after 4-48 h of duloxetine treatment by qRT-PCR. Duloxetine Hydrochloride 79-89 heme oxygenase 1 Homo sapiens 26-30 29134401-12 2018 Finally, the HO-1 inhibitor, ZnPP, suppressed the duloxetine protective effect. Duloxetine Hydrochloride 50-60 heme oxygenase 1 Homo sapiens 13-17 29134401-13 2018 Overall, these results indicate that the mechanism of duloxetine neuroprotective action against oxidative stress and cell death might rely on the Akt/Nrf2/HO-1 pathways. Duloxetine Hydrochloride 54-64 heme oxygenase 1 Homo sapiens 155-159 29496161-9 2018 RESULTS: Three of the nine compounds predicted as Nrf2 activators by molecular docking, gallic acid (GA), Z-liguistilide (LIG), and senkyunolide A (SA), were confirmed with highest potency of increasing the Nrf2/ARE promoter activity and upregulating the expression of Hmox1, Nqo1 and Slc7a11. z-liguistilide 106-120 heme oxygenase 1 Homo sapiens 269-274 30231242-0 2018 Oxysophocarpine Retards the Growth and Metastasis of Oral Squamous Cell Carcinoma by Targeting the Nrf2/HO-1 Axis. oxysophocarpine 0-15 heme oxygenase 1 Homo sapiens 104-108 29361943-0 2018 Identification of danthron as an isoform-specific inhibitor of HEME OXYGENASE-1/cytochrome P450 reductase interaction with anti-tumor activity. danthron 18-26 heme oxygenase 1 Homo sapiens 63-79 29277159-1 2018 The study investigated whether dietary methionine (Met) affects egg weight and antioxidant status through regulating gene expression of ovalbumin (OVAL), nuclear factor erythroid 2 like 2 (Nrf2) and haem oxygenase 1 (HO-1) in laying duck breeders. Methionine 39-49 heme oxygenase 1 Homo sapiens 199-215 29277159-1 2018 The study investigated whether dietary methionine (Met) affects egg weight and antioxidant status through regulating gene expression of ovalbumin (OVAL), nuclear factor erythroid 2 like 2 (Nrf2) and haem oxygenase 1 (HO-1) in laying duck breeders. Methionine 39-49 heme oxygenase 1 Homo sapiens 217-221 30064139-11 2018 Zedoarondiol induced nuclear Nrf2 translocation from cytoplasm into nucleus and up-regulated expression of HO-1, NQO1, and Nrf2 in nucleus. zedoarondiol 0-12 heme oxygenase 1 Homo sapiens 107-111 30064139-13 2018 CONCLUSION: Zedoarondiol attenuates ox-LDL-induced endothelial cells injury by inhibiting oxidative stress and inflammation via Nrf2/HO-1 pathway, suggesting that zedoarondiol might be meaningful on prevention and treatment of atherosclerosis. zedoarondiol 12-24 heme oxygenase 1 Homo sapiens 133-137 30064139-13 2018 CONCLUSION: Zedoarondiol attenuates ox-LDL-induced endothelial cells injury by inhibiting oxidative stress and inflammation via Nrf2/HO-1 pathway, suggesting that zedoarondiol might be meaningful on prevention and treatment of atherosclerosis. zedoarondiol 163-175 heme oxygenase 1 Homo sapiens 133-137 29694447-0 2018 Pharmacological versus genetic inhibition of heme oxygenase-1 - the comparison of metalloporphyrins, shRNA and CRISPR/Cas9 system. Metalloporphyrins 82-99 heme oxygenase 1 Homo sapiens 45-61 29694447-4 2018 Metalloporphyrins decreased HO activity but concomitantly strongly induced HO-1 mRNA and protein in 293T cells. Metalloporphyrins 0-17 heme oxygenase 1 Homo sapiens 75-79 29694447-8 2018 Furthermore, CRISPR/Cas9-mediated HO-1 depletion decreased 293T viability, growth, clonogenic potential and increased sensitivity to H2O2 treatment. Hydrogen Peroxide 133-137 heme oxygenase 1 Homo sapiens 34-38 30068872-9 2018 In contrast, DBM effectively increased the expression of nuclear factor E2-related factor 2 (Nrf2) and its target protein, hemeoxygenase-1 (HO-1). dibenzoylmethane 13-16 heme oxygenase 1 Homo sapiens 123-138 30674243-0 2018 Structural Insights into Azole-based Inhibitors of Heme Oxygenase-1: Development of Selective Compounds for Therapeutic Applications. Azoles 25-30 heme oxygenase 1 Homo sapiens 51-67 30466076-7 2018 RESULTS: We demonstrated that OI, the cell-permeable derivative of itaconate, induced Keap1-Nrf2 dissociation, Nrf2 protein accumulation, and nuclear translocation, which enabled the transcription and expression of multiple Nrf2-dependentantioxidant enzymes (heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1, and glutamate-cysteine ligase modifier subunit) in THP-1 human macrophages. itaconic acid 67-76 heme oxygenase 1 Homo sapiens 259-309 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Polyphenols 20-31 heme oxygenase 1 Homo sapiens 239-255 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Polyphenols 20-31 heme oxygenase 1 Homo sapiens 257-261 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Curcumin 40-48 heme oxygenase 1 Homo sapiens 239-255 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Curcumin 40-48 heme oxygenase 1 Homo sapiens 257-261 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Quercetin 50-59 heme oxygenase 1 Homo sapiens 239-255 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Quercetin 50-59 heme oxygenase 1 Homo sapiens 257-261 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Genistein 61-70 heme oxygenase 1 Homo sapiens 239-255 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). Genistein 61-70 heme oxygenase 1 Homo sapiens 257-261 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). caffeic acid phenethyl ester 76-104 heme oxygenase 1 Homo sapiens 239-255 28618991-11 2018 In particular, some polyphenols such as curcumin, quercetin, genistein, and caffeic acid phenethyl ester are able to potently activate nuclear factor erythroid 2-related factor 2 (Nrf2) and related downstream expression of enzymes such as heme oxygenase-1 (HO-1). caffeic acid phenethyl ester 76-104 heme oxygenase 1 Homo sapiens 257-261 29051011-2 2018 Previously, we reported that heme oxygenase-1 can protect LECs from hydrogen peroxide (H2O2)-induced apoptosis and oxidative stress; however, to the best of our knowledge, these protection mechanisms have not yet been explained. Hydrogen Peroxide 68-85 heme oxygenase 1 Homo sapiens 29-45 30039754-3 2018 Nutraceutical measures which may address these issues include taurine and N-acetylcysteine - which may boost placental H2S production; spirulina and phase 2 inducers of heme oxygenase-1 such as lipoic acid - which may down-regulate placental NADPH oxidase activity; and citrulline, high-dose folate, and dietary nitrate - which by supporting placental nitric oxide production may aid proper placentation and hence prevent placental hypoxia. Thioctic Acid 194-205 heme oxygenase 1 Homo sapiens 169-185 29061583-5 2018 Bioengineered NRF2-siRNA was able to significantly knock down NRF2 mRNA and protein levels in human OS 143B and MG63 cells, and subsequently suppressed the expression of NRF2-regulated oxidative enzymes [heme oxygenase-1 and NAD(P)H:quinone oxidoreductase 1] and elevated intracellular levels of reactive oxygen species. Reactive Oxygen Species 296-319 heme oxygenase 1 Homo sapiens 204-257 29232587-0 2018 Discovery of naphtho[1,2-d]oxazole derivatives as potential anti-HCV agents through inducing heme oxygenase-1 expression. naphtho[1,2-d]oxazole 13-34 heme oxygenase 1 Homo sapiens 93-109 29051011-2 2018 Previously, we reported that heme oxygenase-1 can protect LECs from hydrogen peroxide (H2O2)-induced apoptosis and oxidative stress; however, to the best of our knowledge, these protection mechanisms have not yet been explained. Hydrogen Peroxide 87-91 heme oxygenase 1 Homo sapiens 29-45 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Heme 101-105 heme oxygenase 1 Homo sapiens 0-16 28818727-8 2018 In vivo and in vitro experiments have demonstrated the ability of polysaccharide-rich extracts to provide neuroprotective effects through promotion of neurite outgrowth, and NF-kappaB, PI3K/Akt, MAPK, Nrf2/HO-1 signaling pathways. Polysaccharides 66-80 heme oxygenase 1 Homo sapiens 206-210 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Carbon Monoxide 117-132 heme oxygenase 1 Homo sapiens 63-79 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Carbon Monoxide 117-132 heme oxygenase 1 Homo sapiens 81-85 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Carbon Monoxide 134-136 heme oxygenase 1 Homo sapiens 63-79 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Carbon Monoxide 134-136 heme oxygenase 1 Homo sapiens 81-85 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Biliverdine 142-152 heme oxygenase 1 Homo sapiens 63-79 29791903-1 2018 In mucosal inflammatory disorders, the protective influence of heme oxygenase-1 (HO-1) and its metabolic byproducts, carbon monoxide (CO) and biliverdin, is a topic of significant interest. Biliverdine 142-152 heme oxygenase 1 Homo sapiens 81-85 28823537-5 2018 4-Hydroperoxy-2-decenoic acid ethyl ester (HPO-DAEE), a synthesized RJ fatty acid derivative, markedly induced antioxidant enzymes such as heme oxygenase-1 (HO-1). 4-hydroperoxy-2-decenoic acid ethyl ester 0-41 heme oxygenase 1 Homo sapiens 139-155 28823537-5 2018 4-Hydroperoxy-2-decenoic acid ethyl ester (HPO-DAEE), a synthesized RJ fatty acid derivative, markedly induced antioxidant enzymes such as heme oxygenase-1 (HO-1). hpo-daee 43-51 heme oxygenase 1 Homo sapiens 139-155 28823537-5 2018 4-Hydroperoxy-2-decenoic acid ethyl ester (HPO-DAEE), a synthesized RJ fatty acid derivative, markedly induced antioxidant enzymes such as heme oxygenase-1 (HO-1). Fatty Acids 71-81 heme oxygenase 1 Homo sapiens 139-155 29425644-0 2018 Ligustilide attenuates vascular inflammation and activates Nrf2/HO-1 induction and, NO synthesis in HUVECs. ligustilide 0-11 heme oxygenase 1 Homo sapiens 64-68 29425644-10 2018 Also, ligustilide treated HUVECs exhibited significant HO-1 induction via Nrf2 nuclear translocation and endothelial NO synthesis. ligustilide 6-17 heme oxygenase 1 Homo sapiens 55-59 29029310-9 2018 Moreover, LINA-OOH induced a stronger Nrf2 accumulation in correlation with nqo1 and ho-1 gene expression, 2 Nrf2 target genes. linalool 10-14 heme oxygenase 1 Homo sapiens 85-89 29029310-9 2018 Moreover, LINA-OOH induced a stronger Nrf2 accumulation in correlation with nqo1 and ho-1 gene expression, 2 Nrf2 target genes. OOH 15-18 heme oxygenase 1 Homo sapiens 85-89 29203625-3 2018 Long version of the guanine-thymine (GTn) microsatellite in the HMOX1 promoter decreases HO-1 expression, and the long maternal repeat is associated with late-onset preeclampsia. guanine-thymine (gtn 20-40 heme oxygenase 1 Homo sapiens 64-69 29203625-3 2018 Long version of the guanine-thymine (GTn) microsatellite in the HMOX1 promoter decreases HO-1 expression, and the long maternal repeat is associated with late-onset preeclampsia. guanine-thymine (gtn 20-40 heme oxygenase 1 Homo sapiens 89-93 28083817-0 2018 Carnosic Acid Induces Anti-Inflammatory Effects in Paraquat-Treated SH-SY5Y Cells Through a Mechanism Involving a Crosstalk Between the Nrf2/HO-1 Axis and NF-kappaB. salvin 0-13 heme oxygenase 1 Homo sapiens 141-145 28083817-0 2018 Carnosic Acid Induces Anti-Inflammatory Effects in Paraquat-Treated SH-SY5Y Cells Through a Mechanism Involving a Crosstalk Between the Nrf2/HO-1 Axis and NF-kappaB. Paraquat 51-59 heme oxygenase 1 Homo sapiens 141-145 28083817-6 2018 SH-SY5Y cells were pretreated for 12 h with CA at 1 muM before exposure to PQ for further 24 h. CA suppressed the PQ-induced alterations on the levels of interleukin-1beta (IL-1beta), tumor necrosis factor-alpha (TNF-alpha), and cyclooxygenase-2 (COX-2) through a mechanism involving the activation of the Nrf2/HO-1 axis. Paraquat 114-116 heme oxygenase 1 Homo sapiens 311-315 28083817-7 2018 Furthermore, we observed a crosstalk between the Nrf2/HO-1 signaling pathway and the activation of the nuclear factor-kappaB (NF-kappaB) transcription factor, since administration of ZnPP IX (specific inhibitor of HO-1) or Nrf2 knockdown using small interfering RNA (siRNA) abolished the anti-inflammatory effects induced by CA. zinc protoporphyrin 183-190 heme oxygenase 1 Homo sapiens 54-58 28083817-7 2018 Furthermore, we observed a crosstalk between the Nrf2/HO-1 signaling pathway and the activation of the nuclear factor-kappaB (NF-kappaB) transcription factor, since administration of ZnPP IX (specific inhibitor of HO-1) or Nrf2 knockdown using small interfering RNA (siRNA) abolished the anti-inflammatory effects induced by CA. zinc protoporphyrin 183-190 heme oxygenase 1 Homo sapiens 214-218 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Heme 101-105 heme oxygenase 1 Homo sapiens 18-22 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Iron 116-120 heme oxygenase 1 Homo sapiens 0-16 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Iron 116-120 heme oxygenase 1 Homo sapiens 18-22 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 122-137 heme oxygenase 1 Homo sapiens 0-16 29153608-7 2017 There were increased levels of iNOS, TNF-alpha, HO1, Nrf2, Wfs-1, XBP-1 and spliced XBP-1 observed in response to hemin treatment and the signaling was found to be partly mediated by cofilin. Hemin 114-119 heme oxygenase 1 Homo sapiens 48-51 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 122-137 heme oxygenase 1 Homo sapiens 18-22 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 139-141 heme oxygenase 1 Homo sapiens 0-16 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Carbon Monoxide 139-141 heme oxygenase 1 Homo sapiens 18-22 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Biliverdine 148-158 heme oxygenase 1 Homo sapiens 0-16 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Biliverdine 148-158 heme oxygenase 1 Homo sapiens 18-22 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Bilirubin 197-206 heme oxygenase 1 Homo sapiens 0-16 28756878-1 2017 Heme oxygenase-1 (HO-1) is the enzyme catalyzing the rate-limiting oxidative degradation of cellular heme into free iron, carbon monoxide (CO), and biliverdin, which is then rapidly converted into bilirubin. Bilirubin 197-206 heme oxygenase 1 Homo sapiens 18-22 28756878-2 2017 By means of these catabolic end-products and by removal of pro-oxidant heme, HO-1 exerts antioxidant, antiapoptotic, and immune-modulating effects, leading to overall cytoprotective and beneficial functions in mammalian cells. Heme 71-75 heme oxygenase 1 Homo sapiens 77-81 29216925-9 2017 Docetaxel treatment modestly decreased Bach2 expression and increased HO-1 level in PC3 cells, whereas a modest increase of HO-1 facilitated docetaxel-induced apoptosis. Docetaxel 0-9 heme oxygenase 1 Homo sapiens 70-74 29216925-9 2017 Docetaxel treatment modestly decreased Bach2 expression and increased HO-1 level in PC3 cells, whereas a modest increase of HO-1 facilitated docetaxel-induced apoptosis. Docetaxel 141-150 heme oxygenase 1 Homo sapiens 124-128 29216925-10 2017 Notably, docetaxel-induced miR-193a-5p upregulation, which in turn inhibits Bach2 expression and thus relieves Bach2 repression of HO-1 expression, partly counteracted docetaxel-induced apoptosis, as evidenced by the increased Bcl-2 and decreased Bax expression. Docetaxel 9-18 heme oxygenase 1 Homo sapiens 131-135 29216925-10 2017 Notably, docetaxel-induced miR-193a-5p upregulation, which in turn inhibits Bach2 expression and thus relieves Bach2 repression of HO-1 expression, partly counteracted docetaxel-induced apoptosis, as evidenced by the increased Bcl-2 and decreased Bax expression. Docetaxel 168-177 heme oxygenase 1 Homo sapiens 131-135 29039478-0 2017 Phosphorylation of eIF2alpha suppresses cisplatin-induced p53 activation and apoptosis by attenuating oxidative stress via ATF4-mediated HO-1 expression in human renal proximal tubular cells. Cisplatin 40-49 heme oxygenase 1 Homo sapiens 137-141 28970059-3 2017 The present study investigated the structural requirements of flavonoids to induce Hmox1 in human aortic endothelial cells (HAEC). Flavonoids 62-72 heme oxygenase 1 Homo sapiens 83-88 28749008-0 2017 Propofol post-conditioning alleviates hepatic ischaemia reperfusion injury via BRG1-mediated Nrf2/HO-1 transcriptional activation in human and mice. Propofol 0-8 heme oxygenase 1 Homo sapiens 98-102 28513870-6 2017 We discovered an increased calcium ion concentration and upregulated Runx2, BMP2, Nrf2, HO-1, gamma-GCS, NQO-1, and LC3II/LC3I expressions in the high phosphorous, si-NC and Nrf2-siRNA, and NC groups, compared with the normal control group. Phosphorus 151-162 heme oxygenase 1 Homo sapiens 88-103 28513870-6 2017 We discovered an increased calcium ion concentration and upregulated Runx2, BMP2, Nrf2, HO-1, gamma-GCS, NQO-1, and LC3II/LC3I expressions in the high phosphorous, si-NC and Nrf2-siRNA, and NC groups, compared with the normal control group. Silicon 133-135 heme oxygenase 1 Homo sapiens 88-103 28513870-7 2017 Compared to the high phosphorus and si-NC groups, higher levels of Runx2 and BMP2 but decreased Nrf2, HO-1, gamma-GCS, NQO-1, and LC3II/LC3I expressions were detected in the Nrf2-siRNA group. Silicon 36-38 heme oxygenase 1 Homo sapiens 102-117 28513870-8 2017 The high phosphorus, si-NC and over-expressed Nrf2 experimental groups all had increased Nrf2, NQO-1, HO-1, gamma-GCS, and LC3II/LC3I expressions as well as high numbers of autophagosomes compared with the normal control group. Phosphorus 9-19 heme oxygenase 1 Homo sapiens 102-117 29152653-11 2017 Furthermore, elaidic acid resulted in upregulation of nuclear factor erythroid 2-related factor 2 and downregulation of heme oxygenase 1, which are two key antioxidative factors. elaidic acid 13-25 heme oxygenase 1 Homo sapiens 120-136 29039478-11 2017 Furthermore, cisplatin induced the expression of activating transcription factor 4 (ATF4) and heme oxygenase-1 (HO-1) that were enhanced by Sal003 and reduced by PERK knockdown. Cisplatin 13-22 heme oxygenase 1 Homo sapiens 94-110 29039478-11 2017 Furthermore, cisplatin induced the expression of activating transcription factor 4 (ATF4) and heme oxygenase-1 (HO-1) that were enhanced by Sal003 and reduced by PERK knockdown. Cisplatin 13-22 heme oxygenase 1 Homo sapiens 112-116 29039478-12 2017 Taken together, these results suggest that phosphorylation of eIF2alpha suppresses cisplatin-induced p53 activation and apoptosis by attenuating oxidative stress via ATF4-mediated HO-1 expression in HK-2 cells, as ATF4 expression is usually dependent on the phosphorylation of eIF2alpha and may also transcriptionally induce the expression of HO-1 in response to oxidative stress. Cisplatin 83-92 heme oxygenase 1 Homo sapiens 180-184 29039478-12 2017 Taken together, these results suggest that phosphorylation of eIF2alpha suppresses cisplatin-induced p53 activation and apoptosis by attenuating oxidative stress via ATF4-mediated HO-1 expression in HK-2 cells, as ATF4 expression is usually dependent on the phosphorylation of eIF2alpha and may also transcriptionally induce the expression of HO-1 in response to oxidative stress. Cisplatin 83-92 heme oxygenase 1 Homo sapiens 343-347 28513870-8 2017 The high phosphorus, si-NC and over-expressed Nrf2 experimental groups all had increased Nrf2, NQO-1, HO-1, gamma-GCS, and LC3II/LC3I expressions as well as high numbers of autophagosomes compared with the normal control group. Silicon 21-23 heme oxygenase 1 Homo sapiens 102-117 28156185-7 2017 RESULTS: DMF and MMF induced NQO1 and HO1 gene expression in ex vivo-stimulated PBMCs, DMF being the more potent inducer. Dimethyl Fumarate 9-12 heme oxygenase 1 Homo sapiens 38-41 28156185-7 2017 RESULTS: DMF and MMF induced NQO1 and HO1 gene expression in ex vivo-stimulated PBMCs, DMF being the more potent inducer. mmf 17-20 heme oxygenase 1 Homo sapiens 38-41 28749008-7 2017 Brg1 and Nrf2 siRNA were used to examine the relationship between Brg1 and Nrf2/HO-1 pathways in propofol-mediated effects in a human hepatocyte(L02) hypoxia/reoxygenation(H/R) model. Propofol 97-105 heme oxygenase 1 Homo sapiens 80-84 28749008-15 2017 Propofol post-conditioning alleviates HIRI through BRG1-mediated Nrf2/HO-1 transcriptional activation. Propofol 0-8 heme oxygenase 1 Homo sapiens 70-74 28882558-0 2017 Cinnamaldehyde protects human dental pulp cells against oxidative stress through the Nrf2/HO-1-dependent antioxidant response. cinnamaldehyde 0-14 heme oxygenase 1 Homo sapiens 90-94 29333383-0 2017 Inhibitory Effects of Butein on Adipogenesis through Upregulation of the Nrf2/HO-1 Pathway in 3T3-L1 Adipocytes. butein 22-28 heme oxygenase 1 Homo sapiens 78-82 29333383-5 2017 Butein also increased Nrf2 and HO-1 protein expression in a dose-dependent manner. butein 0-6 heme oxygenase 1 Homo sapiens 31-35 29333383-6 2017 Treatment with zinc protoporphyrin, a specific HO-1 inhibitor, abolished the inhibitory effects of butein on adipogenic transcription factor protein expression. zinc protoporphyrin 15-34 heme oxygenase 1 Homo sapiens 47-51 29333383-6 2017 Treatment with zinc protoporphyrin, a specific HO-1 inhibitor, abolished the inhibitory effects of butein on adipogenic transcription factor protein expression. butein 99-105 heme oxygenase 1 Homo sapiens 47-51 29333383-7 2017 Therefore, butein inhibits adipogenesis, at least partially, through upregulation of the Nrf-2/HO-1 signaling pathway in 3T3-L1 adipocytes. butein 11-17 heme oxygenase 1 Homo sapiens 95-99 29230104-0 2017 Metformin Sensitizes Non-small Cell Lung Cancer Cells to an Epigallocatechin-3-Gallate (EGCG) Treatment by Suppressing the Nrf2/HO-1 Signaling Pathway. epigallocatechin gallate 60-86 heme oxygenase 1 Homo sapiens 128-132 29230104-0 2017 Metformin Sensitizes Non-small Cell Lung Cancer Cells to an Epigallocatechin-3-Gallate (EGCG) Treatment by Suppressing the Nrf2/HO-1 Signaling Pathway. epigallocatechin gallate 88-92 heme oxygenase 1 Homo sapiens 128-132 29230104-3 2017 The NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway is considered to mediate cellular resistance to EGCG. epigallocatechin gallate 124-128 heme oxygenase 1 Homo sapiens 34-50 29230104-3 2017 The NF-E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway is considered to mediate cellular resistance to EGCG. epigallocatechin gallate 124-128 heme oxygenase 1 Homo sapiens 52-56 29230104-6 2017 In this study, metformin inhibited HO-1 expression and augmented the anti-tumor effect of EGCG. Metformin 15-24 heme oxygenase 1 Homo sapiens 35-39 29230104-9 2017 Mechanistically, metformin modulated the EGCG-activated Nrf2/HO-1 pathway through Sirtuin 1 (SIRT1)-dependent deacetylation of Nrf2. Metformin 17-26 heme oxygenase 1 Homo sapiens 61-65 29230104-9 2017 Mechanistically, metformin modulated the EGCG-activated Nrf2/HO-1 pathway through Sirtuin 1 (SIRT1)-dependent deacetylation of Nrf2. epigallocatechin gallate 41-45 heme oxygenase 1 Homo sapiens 61-65 29230104-12 2017 Based on our findings, metformin sensitized NSCLC cells to the EGCG treatment by suppressing the Nrf2/HO-1 signaling pathway. Metformin 23-32 heme oxygenase 1 Homo sapiens 102-106 29230104-12 2017 Based on our findings, metformin sensitized NSCLC cells to the EGCG treatment by suppressing the Nrf2/HO-1 signaling pathway. epigallocatechin gallate 63-67 heme oxygenase 1 Homo sapiens 102-106 29170479-1 2017 Heme oxygenase-1-derived carbon monoxide prevents inflammatory vascular disorders. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 29209333-7 2017 In addition, DMF treatment upregulated antioxidant enzymes heme oxygenase-1 and glutathione S-transferase-alpha1 expressions. Dimethyl Fumarate 13-16 heme oxygenase 1 Homo sapiens 59-112 29230104-0 2017 Metformin Sensitizes Non-small Cell Lung Cancer Cells to an Epigallocatechin-3-Gallate (EGCG) Treatment by Suppressing the Nrf2/HO-1 Signaling Pathway. Metformin 0-9 heme oxygenase 1 Homo sapiens 128-132 28882558-5 2017 CA-mediated Nrf2/HO-1 activation reduced the level of reactive oxygen species and protected the hDPCs from H2O2-induced oxidative stress, which induces apoptosis. Reactive Oxygen Species 54-77 heme oxygenase 1 Homo sapiens 17-21 29022012-11 2017 Vanadium downregulated P38, ERK1/2, JNK, Nrf2, sMaf, GCLC, NQO1 and HO-1 mRNA expression (P < 0.05). Vanadium 0-8 heme oxygenase 1 Homo sapiens 68-72 28882558-5 2017 CA-mediated Nrf2/HO-1 activation reduced the level of reactive oxygen species and protected the hDPCs from H2O2-induced oxidative stress, which induces apoptosis. Hydrogen Peroxide 107-111 heme oxygenase 1 Homo sapiens 17-21 28602161-7 2017 Thus, geraniin up-regulates HO-1 expression both through activating its positive regulator Nrf-2 and by down-regulating its negative regulator BACH-1. Geraniin 6-14 heme oxygenase 1 Homo sapiens 28-32 29125538-3 2017 CYP1A1 is mediated by aryl hydrocarbon receptor signaling, while induction of HO-1 is regulated by oxidative stress, and suppressed by N-acetylcysteine treatment. Acetylcysteine 135-151 heme oxygenase 1 Homo sapiens 78-82 28950658-5 2017 7,8-DHF also significantly reduced the generation of intracellular reactive oxygen species (ROS), induced the expression of anti-oxidant enzymes, such as catalase, manganese superoxide dismutase (Mn-SOD), and heme oxygenase-1 (HO-1), and scavenged DPPH free radicals. 6,7-dihydroxyflavone 0-7 heme oxygenase 1 Homo sapiens 209-225 28950658-5 2017 7,8-DHF also significantly reduced the generation of intracellular reactive oxygen species (ROS), induced the expression of anti-oxidant enzymes, such as catalase, manganese superoxide dismutase (Mn-SOD), and heme oxygenase-1 (HO-1), and scavenged DPPH free radicals. 6,7-dihydroxyflavone 0-7 heme oxygenase 1 Homo sapiens 227-231 28950658-8 2017 7,8-DHF effectively attenuated oxidative stress by up-regulating the anti-oxidant enzymes catalase, Mn-SOD, and HO-1, and reducing activation of the Akt and MAPKs signaling pathways in aged skin cells. 6,7-dihydroxyflavone 0-7 heme oxygenase 1 Homo sapiens 112-116 28602161-8 2017 Up-regulation of HO-1 also confers protection to HepG2 cells from tertiary butyl hydroperoxide (TBH) induced cytotoxicity. n-Butylhydroperoxide 75-94 heme oxygenase 1 Homo sapiens 17-21 28602161-8 2017 Up-regulation of HO-1 also confers protection to HepG2 cells from tertiary butyl hydroperoxide (TBH) induced cytotoxicity. tert-Butylhydroperoxide 96-99 heme oxygenase 1 Homo sapiens 17-21 28732110-0 2017 Growth inhibitory effect of beta-eudesmol on cholangiocarcinoma cells and its potential suppressive effect on heme oxygenase-1 production, STAT1/3 activation, and NF-kappaB downregulation. beta-eudesmol 28-41 heme oxygenase 1 Homo sapiens 110-126 28732110-2 2017 The present study reports for the first time in vitro growth inhibitory activities of beta-eudesmol, the bioactive sesquiterpenoid present in the rhizome of Atractylodes lancea (Thunb) DC., with respect to its underlying potential effects on heme oxygenase-1 (HO-1) production, STAT1/3 phosphorylation, and NF-kappaB protein expression in human CCA cell line CL-6. beta-eudesmol 86-99 heme oxygenase 1 Homo sapiens 242-258 28732110-2 2017 The present study reports for the first time in vitro growth inhibitory activities of beta-eudesmol, the bioactive sesquiterpenoid present in the rhizome of Atractylodes lancea (Thunb) DC., with respect to its underlying potential effects on heme oxygenase-1 (HO-1) production, STAT1/3 phosphorylation, and NF-kappaB protein expression in human CCA cell line CL-6. beta-eudesmol 86-99 heme oxygenase 1 Homo sapiens 260-264 28732110-6 2017 Western blot analysis indicated that beta-eudesmol treatment resulted in significant suppression of HO-1 production in CL-6 cells. beta-eudesmol 37-50 heme oxygenase 1 Homo sapiens 100-104 28732110-8 2017 Taken together, these results suggest that beta-eudesmol exerts significant growth inhibitory activity on CL-6 cells that may be linked to its inhibitory effect on the production of HO-1, phosphorylation of STAT1/3, and expression of major NF-kappaB proteins. beta-eudesmol 43-56 heme oxygenase 1 Homo sapiens 182-186 29122256-7 2017 Macrophages phagocytize the erythrocytes in the extravascular space which stimulates the production of Heme Oxygenase-1 (HO-1) to metabolize the heme. Heme 145-149 heme oxygenase 1 Homo sapiens 103-119 28870684-6 2017 IsoLQ also induced heme oxygenase-1 (HO-1) expression that may be associated with nuclear factor (erythroid-derived 2)-like 2 (Nrf2) nuclear translocation. isoliquiritigenin 0-5 heme oxygenase 1 Homo sapiens 19-35 29122256-7 2017 Macrophages phagocytize the erythrocytes in the extravascular space which stimulates the production of Heme Oxygenase-1 (HO-1) to metabolize the heme. Heme 145-149 heme oxygenase 1 Homo sapiens 121-125 28649040-6 2017 The OPAO assay showed the best correlation with the production of intracellular reactive oxygen species by NCI-H292 cell line assessed by DCFH oxidation and with the expression of anti-oxidant genes (superoxide dismutase 2, heme-oxygenase-1) as well as the proinflammatory response (Interleukin 6) when exposed from 1 to 10 mug/cm2. opao 4-8 heme oxygenase 1 Homo sapiens 224-240 28870684-6 2017 IsoLQ also induced heme oxygenase-1 (HO-1) expression that may be associated with nuclear factor (erythroid-derived 2)-like 2 (Nrf2) nuclear translocation. isoliquiritigenin 0-5 heme oxygenase 1 Homo sapiens 37-41 29048628-3 2017 In this study, the protein expression of HO-1 in human gastric cancer was measured by immunohistochemistry on paraffin sections of 89 paired gastric carcinoma tissues and adjacent non-cancer tissues. Paraffin 110-118 heme oxygenase 1 Homo sapiens 41-45 28901402-0 2017 Carbon monoxide-releasing molecule suppresses inflammatory and osteoclastogenic cytokines in nicotine- and lipopolysaccharide-stimulated human periodontal ligament cells via the heme oxygenase-1 pathway. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 178-194 28901402-0 2017 Carbon monoxide-releasing molecule suppresses inflammatory and osteoclastogenic cytokines in nicotine- and lipopolysaccharide-stimulated human periodontal ligament cells via the heme oxygenase-1 pathway. Nicotine 93-101 heme oxygenase 1 Homo sapiens 178-194 28846887-5 2017 Also, it was demonstrated that nuclear factor erythroid 2-related factor 2 (Nrf2) activation was involved in modulating cellular oxidative stress, where SchB promoted Nrf2 translocation into the nucleus and downstream target gene expression of heme oxygenase-1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1) and Glutamate-cysteine ligase modifier subunit (GCLM). schizandrin B 153-157 heme oxygenase 1 Homo sapiens 244-260 28543094-6 2017 Specific inhibitors for HO-1, HSP90, caspase-3, DNase I and EndoG almost completely blocked the DNA fragmentation induced by graphene exposure. Graphite 125-133 heme oxygenase 1 Homo sapiens 24-28 29048628-5 2017 The effects of decreased HO-1 expression by two strands of small interfered RNAs (siRNAs) on cell apoptosis, proliferation, and invasion of gastric cancer cell lines were examined by flow cytometry, the MTT assay, and the cell migration assay, respectively. monooxyethylene trimethylolpropane tristearate 203-206 heme oxygenase 1 Homo sapiens 25-29 29451663-8 2017 iNOS levels were positively correlated with ISS scores and blood lactic acid values, and HO-1 and endogenous H2S were negatively correlated with ISS scores and blood lactic acid values. Lactic Acid 166-177 heme oxygenase 1 Homo sapiens 89-93 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. ammonium ferrous sulfate 68-72 heme oxygenase 1 Homo sapiens 14-18 29260515-7 2017 10 mumol/L and 20 mumol/L ATO treatment activated BACH1 expression and inhibited miR-155,NQO1 and HO-1 expression,leading to decreased total antioxidant capacity. Arsenic Trioxide 26-29 heme oxygenase 1 Homo sapiens 98-102 29260515-9 2017 Moreover,high expression of miR-155 reduced BACH1 activation and increased NQO1 and HO-1 expression in cells treated with 20 mumol/L ATO ( P<0.05). Arsenic Trioxide 133-136 heme oxygenase 1 Homo sapiens 84-88 29260515-10 2017 CONCLUSION: Restraining total antioxidant capacity contributes to ATO induced cell death,the underlying mechanisms may be that ATO can activate BACH1 expression through inhibition of the miR-155 level,leading to subsequent inhibition of NQO1 and HO-1 expression. Arsenic Trioxide 127-130 heme oxygenase 1 Homo sapiens 246-250 29260515-10 2017 CONCLUSION: Restraining total antioxidant capacity contributes to ATO induced cell death,the underlying mechanisms may be that ATO can activate BACH1 expression through inhibition of the miR-155 level,leading to subsequent inhibition of NQO1 and HO-1 expression. mir-155 187-194 heme oxygenase 1 Homo sapiens 246-250 29163402-1 2017 Heme oxygenase-1 (HO-1) is an inducible enzyme that is expressed in response to physical and chemical stresses, such as ultraviolet radiation, hyperthermia, hypoxia, reactive oxygen species (ROS), as well as cytokines, among others. Reactive Oxygen Species 166-189 heme oxygenase 1 Homo sapiens 0-16 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 14-18 29163402-1 2017 Heme oxygenase-1 (HO-1) is an inducible enzyme that is expressed in response to physical and chemical stresses, such as ultraviolet radiation, hyperthermia, hypoxia, reactive oxygen species (ROS), as well as cytokines, among others. Reactive Oxygen Species 166-189 heme oxygenase 1 Homo sapiens 18-22 29163402-1 2017 Heme oxygenase-1 (HO-1) is an inducible enzyme that is expressed in response to physical and chemical stresses, such as ultraviolet radiation, hyperthermia, hypoxia, reactive oxygen species (ROS), as well as cytokines, among others. Reactive Oxygen Species 191-194 heme oxygenase 1 Homo sapiens 0-16 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Carbon Monoxide 92-94 heme oxygenase 1 Homo sapiens 14-18 29163402-1 2017 Heme oxygenase-1 (HO-1) is an inducible enzyme that is expressed in response to physical and chemical stresses, such as ultraviolet radiation, hyperthermia, hypoxia, reactive oxygen species (ROS), as well as cytokines, among others. Reactive Oxygen Species 191-194 heme oxygenase 1 Homo sapiens 18-22 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Biliverdine 100-110 heme oxygenase 1 Homo sapiens 14-18 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Iron 28-32 heme oxygenase 1 Homo sapiens 14-18 28821394-8 2017 Furthermore, hNSCs exposed to AGEs had significantly lower mRNA levels among other components of normal cellular oxidative defenses (GSH, Catalase and HO-1), which were all rescued by co-treatment with metformin. Metformin 202-211 heme oxygenase 1 Homo sapiens 151-155 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Heme 44-48 heme oxygenase 1 Homo sapiens 14-18 29163402-3 2017 Once induced, HO-1 degrades iron-containing heme into ferrous iron (Fe2+), carbon monoxide (CO) and biliverdin. Iron 54-66 heme oxygenase 1 Homo sapiens 14-18 29085368-5 2017 1,25(OH)2D3 can induce the expression of hemeoxygenase-1 (HO-1), which is essential for blocking HMBG1 nuclear translocation and its secretion. Calcitriol 0-11 heme oxygenase 1 Homo sapiens 41-62 28935193-1 2017 BACKGROUND AND PURPOSE: Celastrol, a quinone methide triterpene isolated from the root extracts of Tripterygium wilfordii, can greatly induce the gene expression activity of heme oxygenase-1 (HO-1) to achieve disease prevention and control. celastrol 24-33 heme oxygenase 1 Homo sapiens 192-196 29056901-9 2017 RSL cells upregulated the expression of NRF2 (an ARE ligand) and NR0B2 in response to CoPP (a HMOX1 inducer), but not to ZnPP (a HMOX1 inhibitor). COPP protocol 86-90 heme oxygenase 1 Homo sapiens 94-99 28935193-10 2017 These antiviral effects were abrogated by treatment with the HO-1-specific inhibitor SnMP or silencing of HO-1 expression by transfection of shRNA, which indicates that HO-1 induction contributes to the anti-HCV activity of celastrol. celastrol 224-233 heme oxygenase 1 Homo sapiens 61-65 28935193-10 2017 These antiviral effects were abrogated by treatment with the HO-1-specific inhibitor SnMP or silencing of HO-1 expression by transfection of shRNA, which indicates that HO-1 induction contributes to the anti-HCV activity of celastrol. celastrol 224-233 heme oxygenase 1 Homo sapiens 106-110 28935193-5 2017 The anti-HCV mechanism of celastrol targeting HO-1 expression was clarified using specific inhibitors against several signaling pathways. celastrol 26-35 heme oxygenase 1 Homo sapiens 46-50 28935193-10 2017 These antiviral effects were abrogated by treatment with the HO-1-specific inhibitor SnMP or silencing of HO-1 expression by transfection of shRNA, which indicates that HO-1 induction contributes to the anti-HCV activity of celastrol. celastrol 224-233 heme oxygenase 1 Homo sapiens 106-110 28935193-11 2017 JNK mitogen-activated protein kinase and nuclear factor erythroid 2-related factor 2 (Nrf2) were confirmed to be involved in the inductive effect of celastrol on HO-1 expression. celastrol 149-158 heme oxygenase 1 Homo sapiens 162-166 28935193-9 2017 Celastrol-induced heme oxygenase 1 (HO-1) expression promoted antiviral interferon responses and inhibition of NS3/4A protease activity, thereby blocking HCV replication. celastrol 0-9 heme oxygenase 1 Homo sapiens 36-40 28935193-10 2017 These antiviral effects were abrogated by treatment with the HO-1-specific inhibitor SnMP or silencing of HO-1 expression by transfection of shRNA, which indicates that HO-1 induction contributes to the anti-HCV activity of celastrol. tin mesoporphyrin 85-89 heme oxygenase 1 Homo sapiens 61-65 28733231-6 2017 Significantly, these results showed that tussilagonone induces Nrf2 activation and nuclear accumulation, resulting in the upregulation of the detoxifying enzymes NAD(P)H quinone dehydrogenase 1(NQO1) and heme oxygenase-1(HO-1) that protect cells from oxidative stress. tussilagonone 41-54 heme oxygenase 1 Homo sapiens 204-225 28757460-5 2017 Meanwhile, olaquindox activated AKT and Nrf2/HO-1 pathway, up-regulated Bax/Bcl-2 ratio, disrupted mitochondrial membrane potential (MMP) and subsequently caused cytochrome c release and a cascade activation of caspase, eventually induced apoptosis. olaquindox 11-21 heme oxygenase 1 Homo sapiens 45-49 28757460-7 2017 Furthermore, knockdown of p21 decreased cell viability, enhanced oxidative stress, aggravated olaquindox-induced mitochondrial apoptosis and S-phase arrest involvement of activating PI3K/AKT and inhibiting Nrf2/HO-1 pathway. olaquindox 94-104 heme oxygenase 1 Homo sapiens 211-215 28791341-10 2017 Inhibition of HO-1 with zinc protoporphyrin significantly abolished apocynin-induced suppression of MCP-1, indicating that HO-1 is significant in the suppression of MCP-1. zinc protoporphyrin 24-43 heme oxygenase 1 Homo sapiens 14-18 28791341-10 2017 Inhibition of HO-1 with zinc protoporphyrin significantly abolished apocynin-induced suppression of MCP-1, indicating that HO-1 is significant in the suppression of MCP-1. acetovanillone 68-76 heme oxygenase 1 Homo sapiens 14-18 28791341-10 2017 Inhibition of HO-1 with zinc protoporphyrin significantly abolished apocynin-induced suppression of MCP-1, indicating that HO-1 is significant in the suppression of MCP-1. acetovanillone 68-76 heme oxygenase 1 Homo sapiens 123-127 28791341-11 2017 Thus, apocynin exerts antiseptic activity via the suppression of pro-inflammatory signaling pathways and the activation of cytoprotective signaling pathways, such as HO-1/Nrf-2 in RMCs, indicating that apocynin may present as a promising candidate for in vivo evaluation of a therapeutic agent for inflammation-associated renal disorders. acetovanillone 6-14 heme oxygenase 1 Homo sapiens 166-170 28583303-10 2017 HO-1, a major gene that is upregulated in RNA-sequencing using Keap1-silenced PHEMs, protected melanocytes against H2O2-induced cell death and upregulated MITF and beta-catenin expression. Hydrogen Peroxide 115-119 heme oxygenase 1 Homo sapiens 0-4 29375050-11 2017 ATB-346 significantly increased Nrf-2 and HO-1 protein expression as compared with vehicle (P < 0.05) but did not affect the protein expression of CSE, CBS, 3-MST or HIF-1alpha. 4-anisyltetrazolium blue 0-3 heme oxygenase 1 Homo sapiens 42-46 28791341-0 2017 Apocynin protects mesangial cells from lipopolysaccharide-induced inflammation by exerting heme oxygenase 1-mediated monocyte chemoattractant protein-1 suppression. acetovanillone 0-8 heme oxygenase 1 Homo sapiens 91-107 29375050-17 2017 The decreased gastrotoxicity of ATB-346 could be due to upregulation of Nrf-2/HO-1 pathway mediated by the release of H2s. 4-anisyltetrazolium blue 32-35 heme oxygenase 1 Homo sapiens 78-82 29375050-17 2017 The decreased gastrotoxicity of ATB-346 could be due to upregulation of Nrf-2/HO-1 pathway mediated by the release of H2s. Hydrogen Sulfide 118-121 heme oxygenase 1 Homo sapiens 78-82 27686076-7 2017 We found that the CA-induced Nrf2-dependent HO-1 upregulation ameliorated, at least in part, the mitochondrial function in PQ-treated cells. Paraquat 123-125 heme oxygenase 1 Homo sapiens 44-48 28782507-0 2017 MAPK/JNK1 activation protects cells against cadmium-induced autophagic cell death via differential regulation of catalase and heme oxygenase-1 in oral cancer cells. Cadmium 44-51 heme oxygenase 1 Homo sapiens 126-142 28782507-2 2017 We investigated the roles of heme oxygenase-1 (HO-1) and catalase in cadmium (Cd)-induced oxidative stress and the underlying molecular mechanism in oral cancer cells. Cadmium 69-76 heme oxygenase 1 Homo sapiens 47-51 28782507-8 2017 Catalase inhibition by pharmacological and genetic modulations increased Cd-induced ROS production, autophagy, and apoptosis, but suppressed HO-1, indicating that catalase is required for HO-1 induction. Cadmium 73-75 heme oxygenase 1 Homo sapiens 188-192 28782507-3 2017 Exposing YD8 cells to Cd reduced the expression levels of catalase and superoxide dismutase 1/2 and induced the expression of HO-1 as well as autophagy and apoptosis, which were reversed by N-acetyl-l-cysteine (NAC). Cadmium 22-24 heme oxygenase 1 Homo sapiens 126-130 28782507-3 2017 Exposing YD8 cells to Cd reduced the expression levels of catalase and superoxide dismutase 1/2 and induced the expression of HO-1 as well as autophagy and apoptosis, which were reversed by N-acetyl-l-cysteine (NAC). Acetylcysteine 190-209 heme oxygenase 1 Homo sapiens 126-130 28782507-5 2017 Autophagy inhibition via pharmacologic and genetic modulations enhanced Cd-induced HO-1 expression, caspase-3 cleavage, and the production of reactive oxygen species (ROS). Cadmium 72-74 heme oxygenase 1 Homo sapiens 83-87 28928455-0 2017 Polydatin inhibits mast cell-mediated allergic inflammation by targeting PI3K/Akt, MAPK, NF-kappaB and Nrf2/HO-1 pathways. polydatin 0-9 heme oxygenase 1 Homo sapiens 108-112 28669729-7 2017 In addition, Nrf2 and its downstream target effectors (HO1, NQO1) were dramatically attenuated in MC3T3-E cells treatment with H2O2, while MAN can significantly increase the expression of Nrf2, HO1 and NQO1. Hydrogen Peroxide 127-131 heme oxygenase 1 Homo sapiens 55-58 28669729-7 2017 In addition, Nrf2 and its downstream target effectors (HO1, NQO1) were dramatically attenuated in MC3T3-E cells treatment with H2O2, while MAN can significantly increase the expression of Nrf2, HO1 and NQO1. Hydrogen Peroxide 127-131 heme oxygenase 1 Homo sapiens 194-197 28912423-9 2017 In conclusion, FAEW counteracted H2O2-induced oxidative stress through KEAP1-NRF2/HO-1 pathway. Hydrogen Peroxide 33-37 heme oxygenase 1 Homo sapiens 82-86 28735729-13 2017 Moreover, Western blotting analysis demonstrated that protein expressions of Nrf-2 and HO-1 were also up-regulated by flavonoids treatment. Flavonoids 118-128 heme oxygenase 1 Homo sapiens 87-91 28735729-14 2017 CONCLUSIONS: These results clearly demonstrate that the MeOH extracts and flavonoids from Korean milk thistle protected HepG2 cells against oxidative damage triggered by t-BHP principally by modulating ROS generation and restoring depleted GSH levels in addition to the increased Nrf-2/HO-1 signaling cascade. Methanol 56-60 heme oxygenase 1 Homo sapiens 286-290 28735729-14 2017 CONCLUSIONS: These results clearly demonstrate that the MeOH extracts and flavonoids from Korean milk thistle protected HepG2 cells against oxidative damage triggered by t-BHP principally by modulating ROS generation and restoring depleted GSH levels in addition to the increased Nrf-2/HO-1 signaling cascade. Flavonoids 74-84 heme oxygenase 1 Homo sapiens 286-290 28912423-6 2017 Transcriptome-wide microarray analysis indicated NRF2/HO-1 as the common target of FAEW and fluoxetine. Fluoxetine 92-102 heme oxygenase 1 Homo sapiens 54-58 28859441-8 2017 Baicalein co-administration up-regulated expression of Nrf2 and HO-1 mRNAs and down-regulated the expression of NF-kappaB mRNA, compared with those in the colistin alone group. baicalein 0-9 heme oxygenase 1 Homo sapiens 64-68 27992083-6 2017 Consistently, the expression of antioxidant genes, including NF-E2-related factor 2 (Nrf2), haem oxygenase-1 (HO-1) and catalase, was enhanced by afzelin treatment. afzelin 146-153 heme oxygenase 1 Homo sapiens 110-114 27992083-9 2017 The enhancement of HO-1 gene expression by afzelin treatment was reduced by Nrf2-siRNA expression. afzelin 43-50 heme oxygenase 1 Homo sapiens 19-23 28596283-9 2017 However, CDDO-Me was also found to increase the expression of heme-oxygenase-1, a heat-shock-protein that triggers drug resistance and cell survival. bardoxolone methyl 9-16 heme oxygenase 1 Homo sapiens 62-78 28596283-10 2017 We therefore combined CDDO-Me with the heme-oxygenase-1 inhibitor SMA-ZnPP, which also resulted in synergistic growth-inhibitory effects. copoly(styrene-maleic acid)-zinc protoporphyrin 66-74 heme oxygenase 1 Homo sapiens 39-55 28684418-4 2017 Ex vivo experiments indicated that Nrf-2 and HO-1 expression is reduced in T2DM versus non-T2DM OA cartilage (0.57-fold Nrf-2 and 0.34-fold HO-1), and prostaglandin E2 (PGE2) release was increased in samples with low HO-1 expression. Dinoprostone 151-167 heme oxygenase 1 Homo sapiens 45-49 28684418-4 2017 Ex vivo experiments indicated that Nrf-2 and HO-1 expression is reduced in T2DM versus non-T2DM OA cartilage (0.57-fold Nrf-2 and 0.34-fold HO-1), and prostaglandin E2 (PGE2) release was increased in samples with low HO-1 expression. Dinoprostone 169-173 heme oxygenase 1 Homo sapiens 45-49 28870911-9 2017 In addition, sunitinib exhibited concentration-dependent induction of oxidative stress genes (heme oxygenase 1 and glutathione transferase A1) through the nuclear factor erythroid 2-related factor 2 pathway. Sunitinib 13-22 heme oxygenase 1 Homo sapiens 94-110 28651232-8 2017 Further, treatment with silymarin post ICH injury increased Nrf-2/HO-1 and thereby improved overall cytoprotection. Silymarin 24-33 heme oxygenase 1 Homo sapiens 66-70 28651232-9 2017 These findings together show that silymarin acts as neuroprotective compound by preventing inflammatory activation and up regulating Nrf-2/HO-1 signaling post ICH injury. Silymarin 34-43 heme oxygenase 1 Homo sapiens 139-143 28552716-7 2017 In vivo, TUDCA treatment increases the expression of Nrf2, Nrf2 stabilizer DJ-1, and Nrf2 downstream target antioxidant enzymes HO-1 and GPx. ursodoxicoltaurine 9-14 heme oxygenase 1 Homo sapiens 128-132 28684418-7 2017 The HO-1 inducer cobalt protoporphyrin IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1beta-treated cells than in NG+IL-1beta-treated cells. Cobalt 17-23 heme oxygenase 1 Homo sapiens 4-8 28684418-7 2017 The HO-1 inducer cobalt protoporphyrin IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1beta-treated cells than in NG+IL-1beta-treated cells. protoporphyrin IX 24-41 heme oxygenase 1 Homo sapiens 4-8 28684418-7 2017 The HO-1 inducer cobalt protoporphyrin IX more efficiently attenuated PGE2 and IL-6 release in HG+IL-1beta-treated cells than in NG+IL-1beta-treated cells. Dinoprostone 70-74 heme oxygenase 1 Homo sapiens 4-8 28732803-0 2017 (-)-7(S)-hydroxymatairesinol protects against tumor necrosis factor-alpha-mediated inflammation response in endothelial cells by blocking the MAPK/NF-kappaB and activating Nrf2/HO-1. (-)-7(s)-hydroxymatairesinol 0-28 heme oxygenase 1 Homo sapiens 177-181 28633807-0 2017 Differential induction of ATF3 and HO-1 in myeloid cells and keratinocytes via Dimethylfumarate or Cyclosporine A. Dimethyl Fumarate 79-95 heme oxygenase 1 Homo sapiens 35-39 28633807-0 2017 Differential induction of ATF3 and HO-1 in myeloid cells and keratinocytes via Dimethylfumarate or Cyclosporine A. Cyclosporine 99-113 heme oxygenase 1 Homo sapiens 35-39 28713890-0 2017 HO-1 alleviates cholesterol-induced oxidative stress through activation of Nrf2/ERK and inhibition of PI3K/AKT pathways in endothelial cells. Cholesterol 16-27 heme oxygenase 1 Homo sapiens 0-4 28713890-4 2017 It was confirmed that cholesterol stimulation upregulated the expression of HO-1 in a time-dependent manner via the activation and translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), activation of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) signaling pathway and increasing intercellular Ca2+ ([Ca2+]i) concentration. Cholesterol 22-33 heme oxygenase 1 Homo sapiens 76-80 28713890-6 2017 It was confirmed that cholesterol-mediated oxidative damage in vascular endothelial cells induced an increase in the expression of HO-1 via the activation of Nrf2 and the MAPK/ERK signaling pathway, and increasing the [Ca2+]i concentration. Cholesterol 22-33 heme oxygenase 1 Homo sapiens 131-135 29152091-0 2017 Fibroblast growth factor-2-mediated protection of cardiomyocytes from the toxic effects of doxorubicin requires the mTOR/Nrf-2/HO-1 pathway. Doxorubicin 91-102 heme oxygenase 1 Homo sapiens 127-131 29152091-7 2017 A selective inhibitor for HO-1, Tin-Protoporphyrin, prevented the FGF-2 protection against cell death. tin protoporphyrin IX 32-50 heme oxygenase 1 Homo sapiens 26-30 29152091-8 2017 The mTOR inhibitor Rapamycin prevented FGF-2 protection, and blocked the FGF-2 effects on Nrf-2, HO-1 and p62/SQSTM1. Sirolimus 19-28 heme oxygenase 1 Homo sapiens 97-101 29152091-9 2017 Conclusions: In an acute setting Hi- or Lo-FGF-2 protect cardiomyocytes against multiple Dox-induced deleterious effects, by a mechanism dependent on preservation of mTOR activity, Nrf-2 levels, and the upregulation of HO-1. Doxorubicin 89-92 heme oxygenase 1 Homo sapiens 219-223 28583762-6 2017 Furthermore, the effect of puerarin was potentially mediated through activating Nrf2/HO-1 antioxidant signaling pathway and inhibiting Abeta1-40-induced phosphorylation of IRE1 and PERK as well as nuclear expression of ATF6alpha. puerarin 27-35 heme oxygenase 1 Homo sapiens 85-89 28780624-8 2017 Moreover, exogenous CORM-2 could attenuate HO-1 expression, while overexpressed heme oxygenase-1 (HO-1) increased intracellular CO production, attenuated Cx43 expression, hemichannel function, and gap junction function on spinal astrocyte membranes. Carbon Monoxide 20-22 heme oxygenase 1 Homo sapiens 43-47 29070980-3 2017 The haptoglobin-CD163-heme oxygenase-1 (HO-1) pathway circumvents heme toxicity through enzymatic degradation of heme and transcription of antioxidant genes. Heme 22-26 heme oxygenase 1 Homo sapiens 40-44 28655775-5 2017 Herein, we demonstrate that HO1 binds to PCBP2, but not to other PCBP family members, namely PCBP1, PCBP3, or PCBP4. pcbp 41-45 heme oxygenase 1 Homo sapiens 28-31 28806787-7 2017 MG132 increased the levels of HO-1 through the Akt, p38, and Nrf2 signaling pathways. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 heme oxygenase 1 Homo sapiens 30-34 28655775-0 2017 The iron chaperone poly(rC)-binding protein 2 forms a metabolon with the heme oxygenase 1/cytochrome P450 reductase complex for heme catabolism and iron transfer. Iron 4-8 heme oxygenase 1 Homo sapiens 73-89 28655775-8 2017 In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Heme 3-7 heme oxygenase 1 Homo sapiens 36-39 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Iron 51-55 heme oxygenase 1 Homo sapiens 93-109 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Iron 51-55 heme oxygenase 1 Homo sapiens 111-114 28655775-8 2017 In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Heme 3-7 heme oxygenase 1 Homo sapiens 80-83 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Heme 59-63 heme oxygenase 1 Homo sapiens 93-109 28655775-8 2017 In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Heme 22-26 heme oxygenase 1 Homo sapiens 36-39 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Heme 59-63 heme oxygenase 1 Homo sapiens 111-114 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Biliverdine 167-177 heme oxygenase 1 Homo sapiens 93-109 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Biliverdine 167-177 heme oxygenase 1 Homo sapiens 111-114 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Carbon Monoxide 179-194 heme oxygenase 1 Homo sapiens 93-109 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Carbon Monoxide 179-194 heme oxygenase 1 Homo sapiens 111-114 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Iron 208-212 heme oxygenase 1 Homo sapiens 93-109 28655775-1 2017 Mammals incorporate a major proportion of absorbed iron as heme, which is catabolized by the heme oxygenase 1 (HO1)-NADPH-cytochrome P450 reductase (CPR) complex into biliverdin, carbon monoxide, and ferrous iron. Iron 208-212 heme oxygenase 1 Homo sapiens 111-114 28655775-8 2017 In heme-loaded cells, heme prompted HO1-CPR complex formation and decreased the HO1/PCBP2 interaction. Heme 22-26 heme oxygenase 1 Homo sapiens 80-83 28655775-9 2017 Furthermore, in vitro reconstitution experiments with purified recombinant proteins indicated that HO1 could bind to PCBP2 in the presence of heme, whereas loading of PCBP2 with ferrous iron caused PCBP2 to lose its affinity for HO1. Heme 142-146 heme oxygenase 1 Homo sapiens 99-102 28978146-9 2017 In addition, eriodictyol promoted the expression of nuclear factor erythroid 2 p45 related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) up-regulated by As2O3. Arsenic Trioxide 151-156 heme oxygenase 1 Homo sapiens 111-127 28790431-2 2017 The role played by the Nrf2/HO-1 system in favoring cell survival of neuroblastoma (NB) cells exposed to hydrogen peroxide (H2O2) has been investigated using undifferentiated or all-trans retinoic acid (ATRA) differentiated SH-SY5Y cells. Hydrogen Peroxide 105-122 heme oxygenase 1 Homo sapiens 28-32 28790431-4 2017 HO-1 silencing decreased undifferentiated cell viability when exposed to H2O2, proving the role of HO-1 in cell survival. Hydrogen Peroxide 73-77 heme oxygenase 1 Homo sapiens 0-4 28790431-5 2017 Conversely, ATRA differentiated cells exposed to H2O2 showed a significantly lower induction of HO-1, and only the supplementation with low doses of bilirubin (0,5-1 muM) restored viability. Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 96-100 28790431-7 2017 Furthermore, Bach1 was displaced from HO-1 promoter in undifferentiated cells exposed to H2O2, enabling the binding of Nrf2. Hydrogen Peroxide 89-93 heme oxygenase 1 Homo sapiens 38-42 28790431-8 2017 On the contrary, in ATRA differentiated cells treated with H2O2, Bach1 displacement was impaired, preventing Nrf2 binding and limiting HO-1 transcription. Hydrogen Peroxide 59-63 heme oxygenase 1 Homo sapiens 135-139 28612103-0 2017 Dihydromyricetin protects human umbilical vein endothelial cells from injury through ERK and Akt mediated Nrf2/HO-1 signaling pathway. dihydromyricetin 0-16 heme oxygenase 1 Homo sapiens 111-115 28612103-9 2017 The anti-oxidative and anti-apoptotic effects of DMY were abrogated by the transfection of Nrf2 siRNAs and HO-1 inhibitor ZnPP. dihydromyricetin 49-52 heme oxygenase 1 Homo sapiens 107-111 28612103-9 2017 The anti-oxidative and anti-apoptotic effects of DMY were abrogated by the transfection of Nrf2 siRNAs and HO-1 inhibitor ZnPP. zinc protoporphyrin 122-126 heme oxygenase 1 Homo sapiens 107-111 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. dihydromyricetin 13-16 heme oxygenase 1 Homo sapiens 41-45 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. dihydromyricetin 13-16 heme oxygenase 1 Homo sapiens 140-144 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 173-180 heme oxygenase 1 Homo sapiens 41-45 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 173-180 heme oxygenase 1 Homo sapiens 140-144 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 184-192 heme oxygenase 1 Homo sapiens 41-45 28612103-10 2017 Furthermore, DMY might activate the Nrf2/HO-1 pathway through activation of the Akt and ERK1/2 pathways, as shown by the inhibition of Nrf2/HO-1 signaling by the inhibitors PD98059 or LY294002 and the transfection of ERK, Akt siRNAs. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 184-192 heme oxygenase 1 Homo sapiens 140-144 28612103-11 2017 In this study, DMY protects HUVECs from ox-LDL-induced oxidative injury by activating Akt and ERK1/2, which subsequently activates Nrf2/HO-1 signaling, thereby up-regulating antioxidant enzymes and anti-apoptotic proteins. dihydromyricetin 15-18 heme oxygenase 1 Homo sapiens 136-140 27992676-6 2017 RESULTS: CoPP administration increased human HO-1 expression and significantly reduced viraemia, although changes correlated with promoter length (Delta0.5log and Delta2log reduction with medium- and short-polymorphism respectively). cobaltiprotoporphyrin 9-13 heme oxygenase 1 Homo sapiens 45-49 28543773-2 2017 We previously found that HO-1 protein expression is reduced in brains of HIV-infected individuals with HIV-associated neurocognitive disorders (HAND) and in HIV-infected macrophages, where this reduction associates with enhanced glutamate release and neurotoxicity. Glutamic Acid 229-238 heme oxygenase 1 Homo sapiens 25-29 28577438-9 2017 In addition, SchB increased the expression of Nrf2 and HO-1 in a concentration-dependent manner. schizandrin B 13-17 heme oxygenase 1 Homo sapiens 55-59 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Heme 61-65 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Heme 61-65 heme oxygenase 1 Homo sapiens 18-22 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Biliverdine 84-94 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Biliverdine 84-94 heme oxygenase 1 Homo sapiens 18-22 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Bilirubin 109-118 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Bilirubin 109-118 heme oxygenase 1 Homo sapiens 18-22 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Iron 185-189 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Iron 185-189 heme oxygenase 1 Homo sapiens 18-22 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. ferric sulfate 191-195 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. ferric sulfate 191-195 heme oxygenase 1 Homo sapiens 18-22 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Carbon Monoxide 201-216 heme oxygenase 1 Homo sapiens 0-16 28206992-2 2017 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme by which heme is catabolized to biliverdin and thence to bilirubin, with the simultaneous release of equimolar quantities of ferrous iron (Fe3+) and carbon monoxide. Carbon Monoxide 201-216 heme oxygenase 1 Homo sapiens 18-22 28206992-3 2017 Polymorphisms of the HO-1 gene promoter may modulate transcriptional activity, thereby augmenting or attenuating HO-1 expression with resultant modulation of the production of bilirubin. Bilirubin 176-185 heme oxygenase 1 Homo sapiens 21-25 28206992-5 2017 In this clinical review, we surveyed the role of HO-1 gene promoter polymorphisms in the control of bilirubin production and further considered their role, if any, in the pathophysiology of neonatal hyperbilirubinemia. Bilirubin 100-109 heme oxygenase 1 Homo sapiens 49-53 28627599-5 2017 We treated the MCF-7 breast cancer cell line with the HMOX-1 inducer hemin and observed that hemin induced HMOX-1 expression and inhibited migration, invasion and ROS generation in transforming growth factor-beta (TGF-beta)-treated MCF-7 cells using quantitative RT-qPCR, western blotting, wound-healing and cell invasion assays as well as fluorescent probe DCFDA. Reactive Oxygen Species 163-166 heme oxygenase 1 Homo sapiens 54-60 28627599-6 2017 Hemin inhibited TGF-beta-induced EMT in the MCF-7 cells, whereas HMOX-1 siRNA attenuated the suppressive effect of hemin as determined by the expression and cellular distribution of selected EMT markers. Hemin 115-120 heme oxygenase 1 Homo sapiens 65-71 28627599-7 2017 In summary, our results revealed that hemin treatment increased HMOX-1 expression and inhibited TGF-beta-induced EMT in MCF-7 cells. Hemin 38-43 heme oxygenase 1 Homo sapiens 64-70 29108243-1 2017 Heme oxygenase-1 (HO-1) can promote tumor growth and reinforce the resistance of diffuse large B-cell lymphoma (DLBCL) cells to chemotherapeutic drug vincristine. Vincristine 150-161 heme oxygenase 1 Homo sapiens 0-16 28432984-6 2017 The main mediator of cellular adaption to oxidative stress, nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes heme oxygenase (decycling) 1 (HMOX1) or NAD(P)H quinone dehydrogenase 1 (NQO1) were strongly increased by CSEaq in a dose-dependent manner. cseaq 238-243 heme oxygenase 1 Homo sapiens 132-160 28432984-6 2017 The main mediator of cellular adaption to oxidative stress, nuclear factor erythroid 2-related factor 2 (NRF2) and its target genes heme oxygenase (decycling) 1 (HMOX1) or NAD(P)H quinone dehydrogenase 1 (NQO1) were strongly increased by CSEaq in a dose-dependent manner. cseaq 238-243 heme oxygenase 1 Homo sapiens 162-167 28214453-11 2017 Simvastatin decreased iNOS, HO-1, HIF-1alpha and CSE whilst it increased eNOS phosphorylation. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 28-32 28738843-9 2017 Besides, heme oxygenase 1 (HO-1) could be activated by Co exposure; ASX treatment significantly inhibited HO-1 activation, suppressing the oxidative stress induced by Co exposure. astaxanthine 68-71 heme oxygenase 1 Homo sapiens 27-31 28738843-9 2017 Besides, heme oxygenase 1 (HO-1) could be activated by Co exposure; ASX treatment significantly inhibited HO-1 activation, suppressing the oxidative stress induced by Co exposure. astaxanthine 68-71 heme oxygenase 1 Homo sapiens 106-110 29108243-1 2017 Heme oxygenase-1 (HO-1) can promote tumor growth and reinforce the resistance of diffuse large B-cell lymphoma (DLBCL) cells to chemotherapeutic drug vincristine. Vincristine 150-161 heme oxygenase 1 Homo sapiens 18-22 29108243-3 2017 Silencing HO-1 gene expression resisted vorinostat-induced apoptosis and arrested cell cycle in the G0/G1 phase of LY-10 cells. Vorinostat 40-50 heme oxygenase 1 Homo sapiens 10-14 28724911-0 2017 Honokiol inhibits c-Met-HO-1 tumor-promoting pathway and its cross-talk with calcineurin inhibitor-mediated renal cancer growth. honokiol 0-8 heme oxygenase 1 Homo sapiens 24-28 28636034-6 2017 However, zinc(ii)-protoporphyrin IX (ZnPPIX), a selective inhibitor of HO-1, restored the GHP-mediated suppression of ROS production and NOX2, TNF-alpha, IL-1beta, IL-6 and iNOS expression. Reactive Oxygen Species 118-121 heme oxygenase 1 Homo sapiens 71-75 28636034-9 2017 Pharmacological inhibition of Akt, ERK1/2, and p38 abrogated GHP-induced nuclear localization of NF-E2-related factor-2 (Nrf2) and the expression of HO-1. ghp 61-64 heme oxygenase 1 Homo sapiens 149-153 28636034-10 2017 In summary, GHP inhibits the LPS-induced inflammatory status through upregulating HO-1 expression via PI3K/Akt, ERK1/2 and p38 signaling pathways in RAW264.7 macrophages. ghp 12-15 heme oxygenase 1 Homo sapiens 82-86 28636034-2 2017 However, whether heme oxygenase-1 (HO-1) is involved in the cytoprotective effect of GHP against the inflammatory status remains unclear. ghp 85-88 heme oxygenase 1 Homo sapiens 17-33 28636034-2 2017 However, whether heme oxygenase-1 (HO-1) is involved in the cytoprotective effect of GHP against the inflammatory status remains unclear. ghp 85-88 heme oxygenase 1 Homo sapiens 35-39 28636034-6 2017 However, zinc(ii)-protoporphyrin IX (ZnPPIX), a selective inhibitor of HO-1, restored the GHP-mediated suppression of ROS production and NOX2, TNF-alpha, IL-1beta, IL-6 and iNOS expression. zinc(ii)-protoporphyrin ix 9-35 heme oxygenase 1 Homo sapiens 71-75 28636034-6 2017 However, zinc(ii)-protoporphyrin IX (ZnPPIX), a selective inhibitor of HO-1, restored the GHP-mediated suppression of ROS production and NOX2, TNF-alpha, IL-1beta, IL-6 and iNOS expression. znppix 37-43 heme oxygenase 1 Homo sapiens 71-75 28636034-6 2017 However, zinc(ii)-protoporphyrin IX (ZnPPIX), a selective inhibitor of HO-1, restored the GHP-mediated suppression of ROS production and NOX2, TNF-alpha, IL-1beta, IL-6 and iNOS expression. ghp 90-93 heme oxygenase 1 Homo sapiens 71-75 28955775-4 2017 The enhanced heme synthesis in Caco-2 cells was more pronounced under the effect of the combination of ALA and SFC than under the effect of ALA alone, as reflected by the induced expression of heme oxygenase 1 (HO-1), as well as a reduced protein level of the transcriptional corepressor Bach1. Heme 13-17 heme oxygenase 1 Homo sapiens 193-209 28955775-4 2017 The enhanced heme synthesis in Caco-2 cells was more pronounced under the effect of the combination of ALA and SFC than under the effect of ALA alone, as reflected by the induced expression of heme oxygenase 1 (HO-1), as well as a reduced protein level of the transcriptional corepressor Bach1. Heme 13-17 heme oxygenase 1 Homo sapiens 211-215 28955775-4 2017 The enhanced heme synthesis in Caco-2 cells was more pronounced under the effect of the combination of ALA and SFC than under the effect of ALA alone, as reflected by the induced expression of heme oxygenase 1 (HO-1), as well as a reduced protein level of the transcriptional corepressor Bach1. 5-amino levulinic acid 103-106 heme oxygenase 1 Homo sapiens 193-209 28955775-4 2017 The enhanced heme synthesis in Caco-2 cells was more pronounced under the effect of the combination of ALA and SFC than under the effect of ALA alone, as reflected by the induced expression of heme oxygenase 1 (HO-1), as well as a reduced protein level of the transcriptional corepressor Bach1. 5-amino levulinic acid 103-106 heme oxygenase 1 Homo sapiens 211-215 28955775-5 2017 Chromatin immunoprecipitation analysis confirmed Bach1 chromatin occupancy at the enhancer regions of HO-1, which were significantly decreased by the addition of ALA and SFC. 5-amino levulinic acid 162-165 heme oxygenase 1 Homo sapiens 102-106 28347842-0 2017 Potential role of heme metabolism in the inducible expression of heme oxygenase-1. Heme 18-22 heme oxygenase 1 Homo sapiens 65-81 28704474-6 2017 In these animals the catalytic activity of heme oxygenase 1 contributed to the release of elemental iron from the protoporphyrin ring of heme within enterocytes, which may then be transported by the strongly expressed ferroportin across the basolateral membrane to the circulation. Iron 100-104 heme oxygenase 1 Homo sapiens 43-59 28704474-6 2017 In these animals the catalytic activity of heme oxygenase 1 contributed to the release of elemental iron from the protoporphyrin ring of heme within enterocytes, which may then be transported by the strongly expressed ferroportin across the basolateral membrane to the circulation. protoporphyrin IX 114-128 heme oxygenase 1 Homo sapiens 43-59 29089150-0 2017 Hemin Reduces HMGB1 Release by UVB in an AMPK/HO-1-dependent Pathway in Human Keratinocytes HaCaT Cells. Hemin 0-5 heme oxygenase 1 Homo sapiens 46-50 29089150-2 2017 We hypothesized that hemin might reduce HMGB1 release through the induction of HO-1 in UVB-induced HaCaTs. Hemin 21-26 heme oxygenase 1 Homo sapiens 79-83 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Heme 11-15 heme oxygenase 1 Homo sapiens 29-33 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Carbon Monoxide 79-94 heme oxygenase 1 Homo sapiens 11-27 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Carbon Monoxide 79-94 heme oxygenase 1 Homo sapiens 29-33 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Carbon Monoxide 96-98 heme oxygenase 1 Homo sapiens 11-27 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Carbon Monoxide 96-98 heme oxygenase 1 Homo sapiens 29-33 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Iron 109-113 heme oxygenase 1 Homo sapiens 11-27 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Iron 109-113 heme oxygenase 1 Homo sapiens 29-33 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 11-27 28347842-2 2017 The enzyme heme oxygenase-1 (HO-1), involved in the degradation of heme, forms carbon monoxide (CO), ferrous iron, and bilirubin in conjunction with biliverdin reductase, and is induced by various stimuli including oxidative stress and heavy metals. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 29-33 28347842-3 2017 We examined the involvement of heme metabolism in the induction of HO-1 by the inducers sulforaphane and sodium arsenite. Heme 31-35 heme oxygenase 1 Homo sapiens 67-71 28347842-3 2017 We examined the involvement of heme metabolism in the induction of HO-1 by the inducers sulforaphane and sodium arsenite. sulforaphane 88-100 heme oxygenase 1 Homo sapiens 67-71 28347842-3 2017 We examined the involvement of heme metabolism in the induction of HO-1 by the inducers sulforaphane and sodium arsenite. sodium arsenite 105-120 heme oxygenase 1 Homo sapiens 67-71 28445830-0 2017 Overexpression of heme oxygenase-1 in bone marrow stromal cells promotes microenvironment-mediated imatinib resistance in chronic myeloid leukemia. Imatinib Mesylate 99-107 heme oxygenase 1 Homo sapiens 18-34 28445830-6 2017 Following co-culture of BMSCs and K562 cells, up-regulating HO-1 expression in bone marrow stromal cells increased the imatinib (IM) resistance of K562 cells, whereas the apoptosis of K562 cells was effectively promoted without BMSCs co-culture. Imatinib Mesylate 119-127 heme oxygenase 1 Homo sapiens 60-64 28347842-4 2017 METHODS: We examined the expression of HO-1 in sulforaphane-, sodium arsenite- and CORM3-treated HEK293T cells, by measuring the transcriptional activity and levels of mRNA and protein. sulforaphane 47-59 heme oxygenase 1 Homo sapiens 39-43 28347842-4 2017 METHODS: We examined the expression of HO-1 in sulforaphane-, sodium arsenite- and CORM3-treated HEK293T cells, by measuring the transcriptional activity and levels of mRNA and protein. sodium arsenite 62-77 heme oxygenase 1 Homo sapiens 39-43 28347842-5 2017 RESULTS: The blockade of heme biosynthesis by succinylacetone and N-methyl protoporphyrin, which are inhibitors of heme biosynthesis, markedly decreased the induction of HO-1. Heme 25-29 heme oxygenase 1 Homo sapiens 170-174 28347842-5 2017 RESULTS: The blockade of heme biosynthesis by succinylacetone and N-methyl protoporphyrin, which are inhibitors of heme biosynthesis, markedly decreased the induction of HO-1. succinylacetone 46-61 heme oxygenase 1 Homo sapiens 170-174 28347842-5 2017 RESULTS: The blockade of heme biosynthesis by succinylacetone and N-methyl protoporphyrin, which are inhibitors of heme biosynthesis, markedly decreased the induction of HO-1. N-methylprotoporphyrin IX 66-89 heme oxygenase 1 Homo sapiens 170-174 28499242-9 2017 GSP also inhibited the over-generation of TGF-beta1 and p-Smad3, as well as enhanced the levels of Smad7, Nrf-2, HO-1, gamma-GCLC and miR-133a/b. gamma-Thio-GTP 0-3 heme oxygenase 1 Homo sapiens 113-129 28347842-5 2017 RESULTS: The blockade of heme biosynthesis by succinylacetone and N-methyl protoporphyrin, which are inhibitors of heme biosynthesis, markedly decreased the induction of HO-1. Heme 115-119 heme oxygenase 1 Homo sapiens 170-174 28347842-7 2017 The cessation of HO-1 induction occurred at the transcriptional and translational levels, and was mediated by the activation of the heme-binding transcriptional repressor Bach1 and translational factor HRI. Heme 132-136 heme oxygenase 1 Homo sapiens 17-21 28347842-9 2017 CONCLUSIONS: We demonstrated the importance of heme metabolism in the stress-inducible expression of HO-1, and also that heme and its degradation products are protective factors for self-defense responses. Heme 47-51 heme oxygenase 1 Homo sapiens 101-105 28347842-10 2017 GENERAL SIGNIFICANCE: The key role of heme metabolism in the stress-inducible expression of HO-1 may promote further studies on heme and its degradation products as protective factors of cellular stresses and iron homeostasis in specialized cells, organs, and whole animal systems. Heme 38-42 heme oxygenase 1 Homo sapiens 92-96 28347842-10 2017 GENERAL SIGNIFICANCE: The key role of heme metabolism in the stress-inducible expression of HO-1 may promote further studies on heme and its degradation products as protective factors of cellular stresses and iron homeostasis in specialized cells, organs, and whole animal systems. Heme 128-132 heme oxygenase 1 Homo sapiens 92-96 28206713-6 2017 ZO-1 and human HO-1 in Caco2 were significantly decreased after treatment with TNF-alpha; and this effect was reduced when coculture with MSCs from bone marrow. caco2 23-28 heme oxygenase 1 Homo sapiens 15-19 28347842-10 2017 GENERAL SIGNIFICANCE: The key role of heme metabolism in the stress-inducible expression of HO-1 may promote further studies on heme and its degradation products as protective factors of cellular stresses and iron homeostasis in specialized cells, organs, and whole animal systems. Iron 209-213 heme oxygenase 1 Homo sapiens 92-96 28206713-8 2017 In contrast, ZO-1 and human HO-1 increased significantly when the damaged Caco2 was treated with HO-MSCs. caco2 74-79 heme oxygenase 1 Homo sapiens 28-32 28245071-7 2017 Inhibition of miR-153 significantly promoted the expression of Nrf2 and heme oxygenase-1 (HO-1). mir-153 14-21 heme oxygenase 1 Homo sapiens 72-88 28473247-0 2017 Oxidized 5-aminosalicylic acid activates Nrf2-HO-1 pathway by covalently binding to Keap1: Implication in anti-inflammatory actions of 5-aminosalicylic acid. Mesalamine 9-30 heme oxygenase 1 Homo sapiens 46-50 28473247-0 2017 Oxidized 5-aminosalicylic acid activates Nrf2-HO-1 pathway by covalently binding to Keap1: Implication in anti-inflammatory actions of 5-aminosalicylic acid. Mesalamine 135-156 heme oxygenase 1 Homo sapiens 46-50 28473247-9 2017 In parallel, rectal administration of 5-ASA increased the level of HO-1 and nuclear Nrf2 in the inflamed colonic tissues, but not in normal colonic tissues. Mesalamine 38-43 heme oxygenase 1 Homo sapiens 67-71 28473247-10 2017 Moreover, oral gavage of sulfasalazine, a colon-specific prodrug of 5-ASA currently used clinically, activated the Nrf2-HO-1 pathway in the colonic tissues where inflammation was in progress, which was not observed when inflammation subsided. Sulfasalazine 25-38 heme oxygenase 1 Homo sapiens 120-124 28473247-10 2017 Moreover, oral gavage of sulfasalazine, a colon-specific prodrug of 5-ASA currently used clinically, activated the Nrf2-HO-1 pathway in the colonic tissues where inflammation was in progress, which was not observed when inflammation subsided. Mesalamine 68-73 heme oxygenase 1 Homo sapiens 120-124 28473247-11 2017 Collectively, our data suggest that Nrf2-HO-1 pathway is involved in the anti-inflammatory pharmacology of 5-ASA, which was likely being exerted exclusively in the inflammatory state. Mesalamine 107-112 heme oxygenase 1 Homo sapiens 41-45 28245071-9 2017 Our study indicates that the inhibition of miR-153 protects neurons against OGD/R-induced injury by regulating Nrf2/HO-1 signaling and suggests a potential therapeutic target for cerebral ischemia/reperfusion injury. mir-153 43-50 heme oxygenase 1 Homo sapiens 116-120 28559202-10 2017 Treatment with RA remarkably increased the Nrf2 protein expression levels in the nucleus, HO-1 and NQO1 protein expression levels, and the activities of SOD and CAT. rosmarinic acid 15-17 heme oxygenase 1 Homo sapiens 90-94 28245071-7 2017 Inhibition of miR-153 significantly promoted the expression of Nrf2 and heme oxygenase-1 (HO-1). mir-153 14-21 heme oxygenase 1 Homo sapiens 90-94 28566367-4 2017 Our results show that after hRSV infection, HO-1 induction with metalloporphyrin cobalt protoporphyrin IX significantly reduces the loss of body weight due to hRSV-induced disease. metalloporphyrin cobalt protoporphyrin ix 64-105 heme oxygenase 1 Homo sapiens 44-48 28790819-14 2017 The KC-GN-mediated protection against OGD/R-induced neurotoxicity was diminished by NRF2 and heme oxygenase-1 gene knockdowns. kc-gn 4-9 heme oxygenase 1 Homo sapiens 93-109 28093052-8 2017 Additionally, CCI-induced mitophagy was shown to be mediated by the oxidative stress-related extracellular signal-regulated kinase (ERK)/nuclear factor-erythroid2-like2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway, which may serve to reduce the apoptosis induced by oxidative stress. CCI 14-17 heme oxygenase 1 Homo sapiens 176-192 28093052-8 2017 Additionally, CCI-induced mitophagy was shown to be mediated by the oxidative stress-related extracellular signal-regulated kinase (ERK)/nuclear factor-erythroid2-like2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway, which may serve to reduce the apoptosis induced by oxidative stress. CCI 14-17 heme oxygenase 1 Homo sapiens 194-198 28093052-9 2017 These results suggest that CCI-induced mitophagy serves to diminish apoptosis-mediated intestinal epithelial cell damage and to improve intestinal permeability, via ERK/Nrf2/HO-1 signaling. CCI 27-30 heme oxygenase 1 Homo sapiens 174-178 28343048-0 2017 Protective effect of CDDO-ethyl amide against high-glucose-induced oxidative injury via the Nrf2/HO-1 pathway. 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid ethyl amide 21-37 heme oxygenase 1 Homo sapiens 97-101 28343048-0 2017 Protective effect of CDDO-ethyl amide against high-glucose-induced oxidative injury via the Nrf2/HO-1 pathway. Glucose 51-58 heme oxygenase 1 Homo sapiens 97-101 28343048-6 2017 The protein expression levels of HO-1 and Nrf2 were measured through Western blot RESULTS: CDDO-EA elicited cytoprotective effects against NP cell apoptosis and ROS accumulation induced by high glucose. CDDO-EA 91-98 heme oxygenase 1 Homo sapiens 33-37 28343048-7 2017 CDDO-EA treatment increased the HO-1 and Nrf2 expression abrogated by HO-1, Nrf2, and mitogen-activated protein kinase inhibitors. CDDO-EA 0-7 heme oxygenase 1 Homo sapiens 32-36 28343048-7 2017 CDDO-EA treatment increased the HO-1 and Nrf2 expression abrogated by HO-1, Nrf2, and mitogen-activated protein kinase inhibitors. CDDO-EA 0-7 heme oxygenase 1 Homo sapiens 70-74 28343048-8 2017 CONCLUSIONS: The phosphorylation and nuclear translocation of Nrf2 are crucial for HO-1 overexpression induced by CDDO-EA, which is essential for the cytoprotection against high-glucose-induced oxidative stress in NP cells. CDDO-EA 114-121 heme oxygenase 1 Homo sapiens 83-87 28343048-8 2017 CONCLUSIONS: The phosphorylation and nuclear translocation of Nrf2 are crucial for HO-1 overexpression induced by CDDO-EA, which is essential for the cytoprotection against high-glucose-induced oxidative stress in NP cells. Glucose 178-185 heme oxygenase 1 Homo sapiens 83-87 28978042-0 2017 Translocation of heme oxygenase-1 contributes to imatinib resistance in chronic myelogenous leukemia. Imatinib Mesylate 49-57 heme oxygenase 1 Homo sapiens 17-33 28978042-1 2017 Heme oxygenase-1 (HO-1) degrades heme to bilirubin. Heme 33-37 heme oxygenase 1 Homo sapiens 0-16 28978042-1 2017 Heme oxygenase-1 (HO-1) degrades heme to bilirubin. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 28978042-1 2017 Heme oxygenase-1 (HO-1) degrades heme to bilirubin. Bilirubin 41-50 heme oxygenase 1 Homo sapiens 0-16 28978042-1 2017 Heme oxygenase-1 (HO-1) degrades heme to bilirubin. Bilirubin 41-50 heme oxygenase 1 Homo sapiens 18-22 28978042-7 2017 Since translocation of HO-1 disrupts the association with cytochrome P450 reductase, heme degrading activity was higher for ER anchored versus anchorless HO-1. Heme 85-89 heme oxygenase 1 Homo sapiens 23-27 28978042-7 2017 Since translocation of HO-1 disrupts the association with cytochrome P450 reductase, heme degrading activity was higher for ER anchored versus anchorless HO-1. Heme 85-89 heme oxygenase 1 Homo sapiens 154-158 28978042-8 2017 Cell viability tests with increasing concentrations of imatinib showed IC50-values for all six cell lines with ER localized HO-1 that were similar to control cells. Imatinib Mesylate 55-63 heme oxygenase 1 Homo sapiens 124-128 28978042-9 2017 However, out of the seven cell lines with anchorless HO-1, two showed a statistically significant increase in the imatinib IC50 (19.76 muM and 12.35 muM versus 2.35 - 7.57 muM of sensitive cell lines) corresponding to plasma concentrations outside the therapeutic range. Imatinib Mesylate 114-122 heme oxygenase 1 Homo sapiens 53-57 28978042-10 2017 We conclude that the presence of translocated HO-1 in the cytosol and nucleus supports imatinib resistance while it is not sufficient to cause imatinib resistance in every cell line. Imatinib Mesylate 87-95 heme oxygenase 1 Homo sapiens 46-50 28978042-11 2017 In contrast, an increase in ER anchored HO-1 with high heme degrading activity does not contribute to imatinib resistance. Heme 55-59 heme oxygenase 1 Homo sapiens 40-44 28500562-12 2017 In GBM cell lines, 5-ALA induced fluorescence is variable and influenced by EGF-induced downstream activation of HO-1. 5-amino levulinic acid 19-24 heme oxygenase 1 Homo sapiens 113-117 28500562-13 2017 HO-1 protein expression was identified as a negative regulator of 5-ALA induced fluorescence in GBM cells. 5-amino levulinic acid 66-71 heme oxygenase 1 Homo sapiens 0-4 28617444-0 2017 Heme oxygenase-1 derived carbon monoxide suppresses Abeta1-42 toxicity in astrocytes. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 28634389-0 2017 Novel indolyl-chalcone derivatives inhibit A549 lung cancer cell growth through activating Nrf-2/HO-1 and inducing apoptosis in vitro and in vivo. indolyl chalcone 6-22 heme oxygenase 1 Homo sapiens 97-101 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 147-162 heme oxygenase 1 Homo sapiens 13-29 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 147-162 heme oxygenase 1 Homo sapiens 31-35 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 147-162 heme oxygenase 1 Homo sapiens 206-210 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 164-166 heme oxygenase 1 Homo sapiens 13-29 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 164-166 heme oxygenase 1 Homo sapiens 31-35 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Carbon Monoxide 164-166 heme oxygenase 1 Homo sapiens 206-210 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Reactive Oxygen Species 311-314 heme oxygenase 1 Homo sapiens 13-29 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). Reactive Oxygen Species 311-314 heme oxygenase 1 Homo sapiens 31-35 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). UNII-042A8N37WH 63-68 heme oxygenase 1 Homo sapiens 13-29 28617444-7 2017 Induction of haem oxygenase-1 (HO-1) protected astrocytes from Abeta(1-42) toxicity, and this protective effect was mimicked by application of the carbon monoxide (CO) releasing molecule CORM-2, suggesting HO-1 protection was attributable to its formation of CO. CO suppressed the rise of NADPH oxidase-derived ROS caused by Abeta(1-42). UNII-042A8N37WH 63-68 heme oxygenase 1 Homo sapiens 31-35 28617444-8 2017 Under hypoxic conditions (0.5% O2, 48 h) HO-1 was induced in astrocytes and Abeta(1-42) toxicity was significantly reduced, an effect which was reversed by the specific HO-1 inhibitor, QC-15. Oxygen 31-33 heme oxygenase 1 Homo sapiens 41-45 28617444-8 2017 Under hypoxic conditions (0.5% O2, 48 h) HO-1 was induced in astrocytes and Abeta(1-42) toxicity was significantly reduced, an effect which was reversed by the specific HO-1 inhibitor, QC-15. Oxygen 31-33 heme oxygenase 1 Homo sapiens 169-173 28617444-8 2017 Under hypoxic conditions (0.5% O2, 48 h) HO-1 was induced in astrocytes and Abeta(1-42) toxicity was significantly reduced, an effect which was reversed by the specific HO-1 inhibitor, QC-15. TERT-BUTYL 6-OXO-2-AZASPIRO[3.3]HEPTANE-2-CARBOXYLATE 185-190 heme oxygenase 1 Homo sapiens 41-45 28617444-8 2017 Under hypoxic conditions (0.5% O2, 48 h) HO-1 was induced in astrocytes and Abeta(1-42) toxicity was significantly reduced, an effect which was reversed by the specific HO-1 inhibitor, QC-15. TERT-BUTYL 6-OXO-2-AZASPIRO[3.3]HEPTANE-2-CARBOXYLATE 185-190 heme oxygenase 1 Homo sapiens 169-173 28604588-0 2017 Docosahexaenoic Acid Induces Expression of Heme Oxygenase-1 and NAD(P)H:quinone Oxidoreductase through Activation of Nrf2 in Human Mammary Epithelial Cells. Docosahexaenoic Acids 0-20 heme oxygenase 1 Homo sapiens 43-59 28604588-2 2017 In this study, we found that DHA induced expression of two representative antioxidant/cytoprotective enzymes, heme oxygenase-1 (HO-1) and NAD(P)H:quinone oxidoreductase (NQO1), in human mammary epithealial (MCF-10A) cells. Docosahexaenoic Acids 29-32 heme oxygenase 1 Homo sapiens 110-126 28604588-9 2017 In conclusion, DHA activates Nrf2, possibly through modification of critical Keap1 cysteine 288 residue and PKCdelta-mediated phosphorylation of Nrf2, leading to upregulation of HO-1 and NQO1 expression. Docosahexaenoic Acids 15-18 heme oxygenase 1 Homo sapiens 178-182 28604588-4 2017 Nrf2 gene silencing by siRNA abolished the DHA-induced expression of HO-1 and NQO1 proteins. Docosahexaenoic Acids 43-46 heme oxygenase 1 Homo sapiens 69-73 28604588-5 2017 When MCF-10A cells were transfected with mutant constructs in which the cysteine 151 or 288 residue of Keap1 was replaced by serine, DHA-induced expression of HO-1 and NQO1 was markedly reduced. Serine 125-131 heme oxygenase 1 Homo sapiens 159-163 28604588-5 2017 When MCF-10A cells were transfected with mutant constructs in which the cysteine 151 or 288 residue of Keap1 was replaced by serine, DHA-induced expression of HO-1 and NQO1 was markedly reduced. Docosahexaenoic Acids 133-136 heme oxygenase 1 Homo sapiens 159-163 28224676-6 2017 RESULTS: Stimulation of hPVECs and human vaginal epithelial cell line, VK2/E6E7 with H2 O2 augmented the expression of Hb-alpha, Hb-beta, Nrf2, heme oxygenase-1 (HO-1), and reactive oxygen species (ROS). Hydrogen Peroxide 85-90 heme oxygenase 1 Homo sapiens 144-160 28598396-0 2017 Embryotoxicity Caused by DON-Induced Oxidative Stress Mediated by Nrf2/HO-1 Pathway. deoxynivalenol 25-28 heme oxygenase 1 Homo sapiens 71-75 28598396-5 2017 At the same time Nrf2/HO-1 pathway is up-regulated by ROS to protect placenta cells from oxidative damage. ros 54-57 heme oxygenase 1 Homo sapiens 22-26 28598396-6 2017 In DON-treated BeWo cells, the level of ROS has time-effect and dose-effect relationships with HO-1 expression. ros 40-43 heme oxygenase 1 Homo sapiens 95-99 28598396-9 2017 Besides, Nrf2/HO-1 pathway accompanied by the "threshold effect" also plays an important role against DON-induced oxidative damage in this process. deoxynivalenol 102-105 heme oxygenase 1 Homo sapiens 14-18 28392465-12 2017 Our study showed that L-carnitine administration activated p38MAPK/Nrf2 signalling, initiating the expression of HO1 and NQO1, which have anti-apoptotic and anti-oxidative effects, respectively. Carnitine 22-33 heme oxygenase 1 Homo sapiens 113-116 28652743-6 2017 Pretreatment of A549 cells on the apical side of the coculture system with the phagocytosis inhibitor cytochalasin B (CB) blocked ZnO-NP-induced HO-1 and PECAM-1 expression in HCAEC, indicating that endocytosis of ZnO-NPs by alveolar epithelial cells was involved in ZnO-NP-induced HO-1 or PECAM-1 expression in endothelial cells. Cytochalasin B 102-116 heme oxygenase 1 Homo sapiens 145-149 28652743-6 2017 Pretreatment of A549 cells on the apical side of the coculture system with the phagocytosis inhibitor cytochalasin B (CB) blocked ZnO-NP-induced HO-1 and PECAM-1 expression in HCAEC, indicating that endocytosis of ZnO-NPs by alveolar epithelial cells was involved in ZnO-NP-induced HO-1 or PECAM-1 expression in endothelial cells. Cytochalasin B 102-116 heme oxygenase 1 Homo sapiens 282-286 28652743-6 2017 Pretreatment of A549 cells on the apical side of the coculture system with the phagocytosis inhibitor cytochalasin B (CB) blocked ZnO-NP-induced HO-1 and PECAM-1 expression in HCAEC, indicating that endocytosis of ZnO-NPs by alveolar epithelial cells was involved in ZnO-NP-induced HO-1 or PECAM-1 expression in endothelial cells. Zinc Oxide 130-133 heme oxygenase 1 Homo sapiens 145-149 28652743-6 2017 Pretreatment of A549 cells on the apical side of the coculture system with the phagocytosis inhibitor cytochalasin B (CB) blocked ZnO-NP-induced HO-1 and PECAM-1 expression in HCAEC, indicating that endocytosis of ZnO-NPs by alveolar epithelial cells was involved in ZnO-NP-induced HO-1 or PECAM-1 expression in endothelial cells. Zinc Oxide 130-133 heme oxygenase 1 Homo sapiens 282-286 28652743-8 2017 ZnO-NP treatment induced lung and systemic inflammation, dyslipidemia, increased levels of serum HO-1 and PECAM-1, and aortic pathological damage. zno-np 0-6 heme oxygenase 1 Homo sapiens 97-101 28224676-6 2017 RESULTS: Stimulation of hPVECs and human vaginal epithelial cell line, VK2/E6E7 with H2 O2 augmented the expression of Hb-alpha, Hb-beta, Nrf2, heme oxygenase-1 (HO-1), and reactive oxygen species (ROS). Reactive Oxygen Species 173-196 heme oxygenase 1 Homo sapiens 144-160 28224676-6 2017 RESULTS: Stimulation of hPVECs and human vaginal epithelial cell line, VK2/E6E7 with H2 O2 augmented the expression of Hb-alpha, Hb-beta, Nrf2, heme oxygenase-1 (HO-1), and reactive oxygen species (ROS). Reactive Oxygen Species 198-201 heme oxygenase 1 Homo sapiens 144-160 28366867-8 2017 5muM Cd2+ exposure significantly augmented nuclear Nrf2, GSH and GCLC, GCLM, HMOX1, TNFalpha and IL-6 mRNA expression but not GSR, however these upsurges were significantly abrogated by ALA or 1muM Cd2+ pre-treatments. Thioctic Acid 186-189 heme oxygenase 1 Homo sapiens 77-82 28596808-7 2017 In addition, signaling pathways involving heme oxygenase-1 and p38 mitogen-activated protein kinase are associated with the alleviative effects of saturated hydrogen saline on LPS-induced autophagy. saturated hydrogen saline 147-172 heme oxygenase 1 Homo sapiens 42-58 28229933-5 2017 We investigated the activation of nuclear factor-E2-related factor 2 (Nrf2) which regulates various antioxidant genes including heme oxygenase-1 (HMOX1), following exposure to sodium arsenite or cadmium chloride in lamin knockdown human cell lines and primary HGPS human fibroblasts. sodium arsenite 176-191 heme oxygenase 1 Homo sapiens 128-144 28675952-4 2017 The expression of three antioxidant proteins, superoxide dismutase (SOD)-2, haem oxygenase (HO)-1, and sirtuin (SIRT) 1, was reduced upon H2O2 stimulation in miR-200c-overexpressed A549 cells. Hydrogen Peroxide 138-142 heme oxygenase 1 Homo sapiens 76-97 28440398-0 2017 Lycopene inhibits NF-kappaB activation and adhesion molecule expression through Nrf2-mediated heme oxygenase-1 in endothelial cells. Lycopene 0-8 heme oxygenase 1 Homo sapiens 94-110 28440398-8 2017 Subsequently, lycopene induced nuclear factor-erythroid 2 related factor 2 (Nrf2) activation, leading to the increased expression of downstream of heme oxygenase-1 (HO-1). Lycopene 14-22 heme oxygenase 1 Homo sapiens 147-163 28440398-8 2017 Subsequently, lycopene induced nuclear factor-erythroid 2 related factor 2 (Nrf2) activation, leading to the increased expression of downstream of heme oxygenase-1 (HO-1). Lycopene 14-22 heme oxygenase 1 Homo sapiens 165-169 28440398-9 2017 The use of siRNA targeting HO-1 blocked the inhibitory effects of lycopene on IkappaB degradation and ICAM-1 expression. Lycopene 66-74 heme oxygenase 1 Homo sapiens 27-31 28440398-10 2017 The inhibitory effects of lycopene thus appear to be mediated through its induction of Nrf2-mediated HO-1 expression. Lycopene 26-34 heme oxygenase 1 Homo sapiens 101-105 28440398-11 2017 Therefore, the findings of the present study indicate that lycopene suppresses the activation of TNFalpha-induced signaling pathways through the upregulation of Nrf2-mediated HO-1 expression. Lycopene 59-67 heme oxygenase 1 Homo sapiens 175-179 28542559-5 2017 Inhibition of ERK5 by siRNA or BIX02189 (a specific ERK5 inhibitor) reduced the statin-induced upregulations of Nrf2, NQO1, and HO-1. BIX 02189 31-39 heme oxygenase 1 Homo sapiens 128-132 28534853-4 2017 Results from in vitro experiments show that 1T3O effectively inhibits 6-hydroxydopamine-induced (6-OHDA-induced) activation of both p38 mitogen-activated protein kinase (MAPK) and caspase-3 in SH-SY5Y cells; and enhances nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Oxidopamine 70-87 heme oxygenase 1 Homo sapiens 276-292 28534853-4 2017 Results from in vitro experiments show that 1T3O effectively inhibits 6-hydroxydopamine-induced (6-OHDA-induced) activation of both p38 mitogen-activated protein kinase (MAPK) and caspase-3 in SH-SY5Y cells; and enhances nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Oxidopamine 70-87 heme oxygenase 1 Homo sapiens 294-298 28534853-4 2017 Results from in vitro experiments show that 1T3O effectively inhibits 6-hydroxydopamine-induced (6-OHDA-induced) activation of both p38 mitogen-activated protein kinase (MAPK) and caspase-3 in SH-SY5Y cells; and enhances nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Oxidopamine 97-103 heme oxygenase 1 Homo sapiens 276-292 28534853-4 2017 Results from in vitro experiments show that 1T3O effectively inhibits 6-hydroxydopamine-induced (6-OHDA-induced) activation of both p38 mitogen-activated protein kinase (MAPK) and caspase-3 in SH-SY5Y cells; and enhances nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expression via phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt) signaling. Oxidopamine 97-103 heme oxygenase 1 Homo sapiens 294-298 28513573-5 2017 Oxidative molecules were altered after the H2O2 exposure, whereby the signaling of Nrf2 was activated with an increase in its downstream effectors, heme oxygenase-1 (HO-1), NAD(P)H:quinone oxidoreductase 1 (NQO1) and gamma-glutamyl cysteine synthetase (gamma-GCS). Hydrogen Peroxide 43-47 heme oxygenase 1 Homo sapiens 148-164 28553288-8 2017 We found that Mtb-induced HO-1 expression requires NADPH oxidase-dependent reactive oxygen species production induced by the early-secreted antigen ESAT-6, which in turn triggers nuclear translocation of the transcription factor NRF-2. Reactive Oxygen Species 75-98 heme oxygenase 1 Homo sapiens 26-30 28500309-7 2017 In HUVECs, resveratrol significantly increased mRNA of anti-oxidant enzymes HO-1, NQO1, GCLC and TXN but did not significantly alter HO-1 protein expression, whilst reducing HO-1 protein in trophoblast. Resveratrol 11-22 heme oxygenase 1 Homo sapiens 76-80 27960551-0 2017 Date fruits inhibit hepatocyte apoptosis and modulate the expression of hepatocyte growth factor, cytochrome P450 2E1 and heme oxygenase-1 in carbon tetrachloride-induced liver fibrosis. Carbon Tetrachloride 142-162 heme oxygenase 1 Homo sapiens 122-138 28229933-5 2017 We investigated the activation of nuclear factor-E2-related factor 2 (Nrf2) which regulates various antioxidant genes including heme oxygenase-1 (HMOX1), following exposure to sodium arsenite or cadmium chloride in lamin knockdown human cell lines and primary HGPS human fibroblasts. sodium arsenite 176-191 heme oxygenase 1 Homo sapiens 146-151 28229933-5 2017 We investigated the activation of nuclear factor-E2-related factor 2 (Nrf2) which regulates various antioxidant genes including heme oxygenase-1 (HMOX1), following exposure to sodium arsenite or cadmium chloride in lamin knockdown human cell lines and primary HGPS human fibroblasts. Cadmium Chloride 195-211 heme oxygenase 1 Homo sapiens 146-151 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 28243792-0 2017 HMOX1 as a marker of iron excess-induced adipose tissue dysfunction, affecting glucose uptake and respiratory capacity in human adipocytes. Iron 21-25 heme oxygenase 1 Homo sapiens 0-5 28243792-0 2017 HMOX1 as a marker of iron excess-induced adipose tissue dysfunction, affecting glucose uptake and respiratory capacity in human adipocytes. Glucose 79-86 heme oxygenase 1 Homo sapiens 0-5 28243792-2 2017 Here, we aimed to investigate the possible role of haem oxygenase 1 (HMOX1) in iron excess-induced adipose tissue dysfunction. Iron 79-83 heme oxygenase 1 Homo sapiens 51-67 28243792-2 2017 Here, we aimed to investigate the possible role of haem oxygenase 1 (HMOX1) in iron excess-induced adipose tissue dysfunction. Iron 79-83 heme oxygenase 1 Homo sapiens 69-74 28243792-5 2017 RESULTS: Adipose tissue HMOX1 was increased in obese participants (p = 0.01) and positively associated with obesity-related metabolic disturbances, and markers of iron accumulation, inflammation and oxidative stress (p < 0.01). Iron 163-167 heme oxygenase 1 Homo sapiens 24-29 28243792-10 2017 In human adipocytes, iron excess and inflammation led to increased HMOX1 mRNA levels. Iron 21-25 heme oxygenase 1 Homo sapiens 67-72 28243792-11 2017 HMOX1 induction (by haem arginate [hemin] administration), resulted in a significant reduction of mitochondrial respiratory capacity (including basal respiration and spare respiratory capacity), glucose uptake and adipogenesis in parallel with increased expression of inflammatory- and iron excess-related genes. heme arginate 20-33 heme oxygenase 1 Homo sapiens 0-5 28243792-11 2017 HMOX1 induction (by haem arginate [hemin] administration), resulted in a significant reduction of mitochondrial respiratory capacity (including basal respiration and spare respiratory capacity), glucose uptake and adipogenesis in parallel with increased expression of inflammatory- and iron excess-related genes. heme arginate 35-40 heme oxygenase 1 Homo sapiens 0-5 28243792-11 2017 HMOX1 induction (by haem arginate [hemin] administration), resulted in a significant reduction of mitochondrial respiratory capacity (including basal respiration and spare respiratory capacity), glucose uptake and adipogenesis in parallel with increased expression of inflammatory- and iron excess-related genes. Glucose 195-202 heme oxygenase 1 Homo sapiens 0-5 28243792-11 2017 HMOX1 induction (by haem arginate [hemin] administration), resulted in a significant reduction of mitochondrial respiratory capacity (including basal respiration and spare respiratory capacity), glucose uptake and adipogenesis in parallel with increased expression of inflammatory- and iron excess-related genes. Iron 286-290 heme oxygenase 1 Homo sapiens 0-5 28243792-12 2017 CONCLUSIONS/INTERPRETATION: HMOX1 is an important marker of iron excess-induced adipose tissue dysfunction and metabolic disturbances in human obesity. Iron 60-64 heme oxygenase 1 Homo sapiens 28-33 28440458-7 2017 Heme oxygenase-1 (HO-1; a microsomal enzyme) expression was significantly decreased while the reactive oxygen species (ROS) level was increased in the CBS siRNA-transfected SMMC-7721 cells. smmc 173-177 heme oxygenase 1 Homo sapiens 0-16 28276576-2 2017 Our group reported decreased risk for ET in carriers of the minor alleles of the rs2071746 and rs1051308 SNPs in the haem-oxygenases 1 and 2 (HMOX1 and HMOX2), respectively, involved in haem metabolism. Heme 117-121 heme oxygenase 1 Homo sapiens 142-147 28212891-9 2017 Although SFN promotes Nrf2 accumulation to upregulate cytoprotective genes (e.g., heme oxygenase-1 and thioredoxin-1), downregulation of endogenous Nrf2 by target-specific siRNA reveals an Nrf2-independent effect for SFN-mediated inhibition of mTOR/p70S6K/S6 signaling and suppression of VSMC proliferation. sulforaphane 9-12 heme oxygenase 1 Homo sapiens 82-98 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Biliverdine 61-71 heme oxygenase 1 Homo sapiens 0-16 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Biliverdine 61-71 heme oxygenase 1 Homo sapiens 18-22 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Iron 78-82 heme oxygenase 1 Homo sapiens 0-16 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Iron 78-82 heme oxygenase 1 Homo sapiens 18-22 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 0-16 28186648-1 2017 Heme oxygenase-1 (HO-1) catalyses the degradation of heme to biliverdin, free iron, and carbon monoxide. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 18-22 29050217-4 2017 The drug efflux transporter P-glycoprotein (P-gp) and the intracellular antioxidant systems, involving Glutathione S-Transferase pi, Glutathione and heme oxygenase-1, were also inhibited via the ROS-depressed Akt/NF-E2-related factor 2 pathway. Reactive Oxygen Species 195-198 heme oxygenase 1 Homo sapiens 149-165 28445528-9 2017 On the other hand, CO production by RuCO (a CO donor) ameliorated PA-induced ER stress, suggesting that CO production followed by HO-1 induction may contribute to the protective effects of baicalein against PA-induced beta-cell dysfunction. Palmitates 66-68 heme oxygenase 1 Homo sapiens 130-134 28445528-9 2017 On the other hand, CO production by RuCO (a CO donor) ameliorated PA-induced ER stress, suggesting that CO production followed by HO-1 induction may contribute to the protective effects of baicalein against PA-induced beta-cell dysfunction. baicalein 189-198 heme oxygenase 1 Homo sapiens 130-134 28445528-9 2017 On the other hand, CO production by RuCO (a CO donor) ameliorated PA-induced ER stress, suggesting that CO production followed by HO-1 induction may contribute to the protective effects of baicalein against PA-induced beta-cell dysfunction. Palmitates 207-209 heme oxygenase 1 Homo sapiens 130-134 28420988-8 2017 Second, heme oxygenases (HOs), in particular the inducible HO isozyme, HO-1, can provide antioxidant cytoprotection via enzymatic degradation of intracellular heme. Heme 8-12 heme oxygenase 1 Homo sapiens 71-75 28286125-0 2017 n-Propyl gallate suppresses lipopolysaccharide-induced inducible nitric oxide synthase activation through protein kinase Cdelta-mediated up-regulation of heme oxygenase-1 in RAW264.7 macrophages. Propyl Gallate 0-16 heme oxygenase 1 Homo sapiens 154-170 28286125-3 2017 In the present study, we showed that n-propyl gallate increases the expression and activity of the heme oxygenase-1 (HO-1), a stress-inducible protein with potent anti-inflammatory activity, in RAW264.7 macrophages. Propyl Gallate 37-53 heme oxygenase 1 Homo sapiens 99-115 28286125-3 2017 In the present study, we showed that n-propyl gallate increases the expression and activity of the heme oxygenase-1 (HO-1), a stress-inducible protein with potent anti-inflammatory activity, in RAW264.7 macrophages. Propyl Gallate 37-53 heme oxygenase 1 Homo sapiens 117-121 28286125-4 2017 The inhibition of the HO-1 activity by treatment with zinc (II) protoporphyrin IX (ZnPP) or by knockdown of the HO-1 expression with small interference RNA significantly reversed the inhibitory effect of n-Propyl gallate on activations of nuclear factor-kappaB (NF-kappaB) and inducible nitric oxide synthase (iNOS) induced by lipopolysaccharide (LPS). protoporphyrin IX 64-81 heme oxygenase 1 Homo sapiens 22-26 28286125-4 2017 The inhibition of the HO-1 activity by treatment with zinc (II) protoporphyrin IX (ZnPP) or by knockdown of the HO-1 expression with small interference RNA significantly reversed the inhibitory effect of n-Propyl gallate on activations of nuclear factor-kappaB (NF-kappaB) and inducible nitric oxide synthase (iNOS) induced by lipopolysaccharide (LPS). zinc protoporphyrin 83-87 heme oxygenase 1 Homo sapiens 22-26 28286125-4 2017 The inhibition of the HO-1 activity by treatment with zinc (II) protoporphyrin IX (ZnPP) or by knockdown of the HO-1 expression with small interference RNA significantly reversed the inhibitory effect of n-Propyl gallate on activations of nuclear factor-kappaB (NF-kappaB) and inducible nitric oxide synthase (iNOS) induced by lipopolysaccharide (LPS). Propyl Gallate 204-220 heme oxygenase 1 Homo sapiens 22-26 28286125-4 2017 The inhibition of the HO-1 activity by treatment with zinc (II) protoporphyrin IX (ZnPP) or by knockdown of the HO-1 expression with small interference RNA significantly reversed the inhibitory effect of n-Propyl gallate on activations of nuclear factor-kappaB (NF-kappaB) and inducible nitric oxide synthase (iNOS) induced by lipopolysaccharide (LPS). Propyl Gallate 204-220 heme oxygenase 1 Homo sapiens 112-116 28286125-5 2017 An additional mechanism study using inhibitors of signaling kinases revealed the involvement of protein kinase Cdelta (PKCdelta) in the expression of HO-1 induced by n-Propyl gallate. Propyl Gallate 166-182 heme oxygenase 1 Homo sapiens 150-154 28286125-8 2017 Taken together, these findings provide the first evidence that n-Propyl gallate exerts its anti-inflammatory effect through PKCdelta-mediated up-regulation of HO-1 in macrophages. Propyl Gallate 63-79 heme oxygenase 1 Homo sapiens 159-163 28139396-7 2017 These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). Iron 114-118 heme oxygenase 1 Homo sapiens 28-32 28421060-3 2017 HO-1 catalyzes the rate-limiting step in the conversion of heme into iron, biliverdin and the gasotransmitter carbon monoxide (CO), all of which share anti-apoptotic, anti-inflammatory, pro-survival, and tumorigenic activities. Heme 59-63 heme oxygenase 1 Homo sapiens 0-4 28421060-3 2017 HO-1 catalyzes the rate-limiting step in the conversion of heme into iron, biliverdin and the gasotransmitter carbon monoxide (CO), all of which share anti-apoptotic, anti-inflammatory, pro-survival, and tumorigenic activities. Iron 69-73 heme oxygenase 1 Homo sapiens 0-4 28421060-3 2017 HO-1 catalyzes the rate-limiting step in the conversion of heme into iron, biliverdin and the gasotransmitter carbon monoxide (CO), all of which share anti-apoptotic, anti-inflammatory, pro-survival, and tumorigenic activities. Biliverdine 75-85 heme oxygenase 1 Homo sapiens 0-4 28421060-3 2017 HO-1 catalyzes the rate-limiting step in the conversion of heme into iron, biliverdin and the gasotransmitter carbon monoxide (CO), all of which share anti-apoptotic, anti-inflammatory, pro-survival, and tumorigenic activities. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 0-4 28421060-3 2017 HO-1 catalyzes the rate-limiting step in the conversion of heme into iron, biliverdin and the gasotransmitter carbon monoxide (CO), all of which share anti-apoptotic, anti-inflammatory, pro-survival, and tumorigenic activities. Carbon Monoxide 127-129 heme oxygenase 1 Homo sapiens 0-4 27995330-0 2017 Piceatannol attenuates homocysteine-induced endoplasmic reticulum stress and endothelial cell damage via heme oxygenase-1 expression. 3,3',4,5'-tetrahydroxystilbene 0-11 heme oxygenase 1 Homo sapiens 105-121 27995330-0 2017 Piceatannol attenuates homocysteine-induced endoplasmic reticulum stress and endothelial cell damage via heme oxygenase-1 expression. Homocysteine 23-35 heme oxygenase 1 Homo sapiens 105-121 27995330-8 2017 Interestingly, the inhibitory effects of Pic on Hcy-induced apoptosis, ROS generation and ER stress were abolished by down-regulation of HO-1 expression, while mimicked by treatment of ECs with the HO-1 inducer hemin. 3,3',4,5'-tetrahydroxystilbene 41-44 heme oxygenase 1 Homo sapiens 137-141 27995330-8 2017 Interestingly, the inhibitory effects of Pic on Hcy-induced apoptosis, ROS generation and ER stress were abolished by down-regulation of HO-1 expression, while mimicked by treatment of ECs with the HO-1 inducer hemin. 3,3',4,5'-tetrahydroxystilbene 41-44 heme oxygenase 1 Homo sapiens 198-202 27995330-8 2017 Interestingly, the inhibitory effects of Pic on Hcy-induced apoptosis, ROS generation and ER stress were abolished by down-regulation of HO-1 expression, while mimicked by treatment of ECs with the HO-1 inducer hemin. Homocysteine 48-51 heme oxygenase 1 Homo sapiens 137-141 27995330-8 2017 Interestingly, the inhibitory effects of Pic on Hcy-induced apoptosis, ROS generation and ER stress were abolished by down-regulation of HO-1 expression, while mimicked by treatment of ECs with the HO-1 inducer hemin. Homocysteine 48-51 heme oxygenase 1 Homo sapiens 198-202 27995330-9 2017 Overall, these results suggest that Pic may protect ECs against Hcy-induced apoptosis, oxidative stress and ER stress via Nrf2-dependent HO-1 expression. 3,3',4,5'-tetrahydroxystilbene 36-39 heme oxygenase 1 Homo sapiens 137-141 27995330-9 2017 Overall, these results suggest that Pic may protect ECs against Hcy-induced apoptosis, oxidative stress and ER stress via Nrf2-dependent HO-1 expression. Homocysteine 64-67 heme oxygenase 1 Homo sapiens 137-141 28139396-7 2017 These beneficial effects of HO-1 induction during AKI are mediated in part by the by-products of the HO reaction (iron, carbon monoxide, and bile pigments). Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 28-32 28089584-5 2017 Moreover, the levels of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1) and glutathione S-transferase (GST) were elevated after PTE treatment. pterostilbene 156-159 heme oxygenase 1 Homo sapiens 76-92 28213291-5 2017 Furthermore, 2"-O-GH stimulation resulted in a fast and dramatic activation of Akt and nuclear translocation of the NF-E2-related factor 2 (Nrf2), along with the increased expression of heme oxygenase-1 (HO-1) and levels of glutathione (GSH). 2"-o-gh 13-20 heme oxygenase 1 Homo sapiens 186-202 28100868-8 2017 In contrast, retinol treatment significantly increased the mRNA expression of endoplasmic reticulum (ER) stress factors (heme oxygenase 1 (HMOX1), CCAAT/enhancer-binding protein homologous protein (CHOP), 78 kDa glucose-regulated protein (GRP78), and DnaJ (Hsp40) homolog, subfamily B, member 9 (DNAJB9)). Vitamin A 13-20 heme oxygenase 1 Homo sapiens 121-137 28100868-8 2017 In contrast, retinol treatment significantly increased the mRNA expression of endoplasmic reticulum (ER) stress factors (heme oxygenase 1 (HMOX1), CCAAT/enhancer-binding protein homologous protein (CHOP), 78 kDa glucose-regulated protein (GRP78), and DnaJ (Hsp40) homolog, subfamily B, member 9 (DNAJB9)). Vitamin A 13-20 heme oxygenase 1 Homo sapiens 139-144 28103505-0 2017 Hesperetin protects against H2O2-triggered oxidative damage via upregulation of the Keap1-Nrf2/HO-1 signal pathway in ARPE-19 cells. hesperetin 0-10 heme oxygenase 1 Homo sapiens 95-99 28103505-0 2017 Hesperetin protects against H2O2-triggered oxidative damage via upregulation of the Keap1-Nrf2/HO-1 signal pathway in ARPE-19 cells. Hydrogen Peroxide 28-32 heme oxygenase 1 Homo sapiens 95-99 28274716-0 2017 Atractylenolide I restores HO-1 expression and inhibits Ox-LDL-induced VSMCs proliferation, migration and inflammatory responses in vitro. (+)-Atractylenolide 0-15 heme oxygenase 1 Homo sapiens 27-31 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. zinc protoporphyrin 65-84 heme oxygenase 1 Homo sapiens 31-47 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. zinc protoporphyrin 65-84 heme oxygenase 1 Homo sapiens 49-53 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. zinc protoporphyrin 86-90 heme oxygenase 1 Homo sapiens 31-47 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. zinc protoporphyrin 86-90 heme oxygenase 1 Homo sapiens 49-53 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. (+)-Atractylenolide 141-156 heme oxygenase 1 Homo sapiens 31-47 28274716-5 2017 More importantly, the specific heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin (ZnPP) IX partially abolished the beneficial effects of atractylenolide I on Ox-LDL-induced VSMCs. (+)-Atractylenolide 141-156 heme oxygenase 1 Homo sapiens 49-53 28259922-4 2017 HK-2 human tubular epithelial cells exposed to H/R injury were pretreated with LXA4, signal molecule inhibitors or the HO-1 inhibitor zinc protoporphyrin-IX, or were transfected with PPARgamma small interfering RNA (siRNA) or nuclear factor E2-related factor 2 (Nrf2) siRNA. zinc protoporphyrin 134-156 heme oxygenase 1 Homo sapiens 119-123 28070834-0 2017 Resveratrol modulates GSH system in C6 astroglial cells through heme oxygenase 1 pathway. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 64-80 28070834-0 2017 Resveratrol modulates GSH system in C6 astroglial cells through heme oxygenase 1 pathway. Glutathione 22-25 heme oxygenase 1 Homo sapiens 64-80 28070834-7 2017 Additionally, we observed that pharmacological inhibition of heme oxygenase 1 (HO-1), an essential cellular defense against oxidative and inflammatory injuries, abolished all the protective effects of resveratrol. Resveratrol 201-212 heme oxygenase 1 Homo sapiens 61-77 28070834-7 2017 Additionally, we observed that pharmacological inhibition of heme oxygenase 1 (HO-1), an essential cellular defense against oxidative and inflammatory injuries, abolished all the protective effects of resveratrol. Resveratrol 201-212 heme oxygenase 1 Homo sapiens 79-83 28070834-8 2017 These observations suggest HO-1 pathway as a cellular effector in the mechanism by which resveratrol protects astroglial cells against GSH depletion, a condition that may be associated to neurodegenerative diseases. Resveratrol 89-100 heme oxygenase 1 Homo sapiens 27-31 28070834-8 2017 These observations suggest HO-1 pathway as a cellular effector in the mechanism by which resveratrol protects astroglial cells against GSH depletion, a condition that may be associated to neurodegenerative diseases. Glutathione 135-138 heme oxygenase 1 Homo sapiens 27-31 28259922-15 2017 Therefore, LXA4-induced renoprotection is associated with activation of the p38 MAPK/PPARgamma/Nrf2-ARE/HO-1 pathway. lipoxin A4 11-15 heme oxygenase 1 Homo sapiens 104-108 28480030-1 2017 Carbon monoxide (CO) formed endogenously is considered to be cytoprotective, and the vast majority of CO formation is attributed to the degradation of heme by heme oxygenases-1 and -2 (HO-1, HO-2). Heme 151-155 heme oxygenase 1 Homo sapiens 159-183 28480030-1 2017 Carbon monoxide (CO) formed endogenously is considered to be cytoprotective, and the vast majority of CO formation is attributed to the degradation of heme by heme oxygenases-1 and -2 (HO-1, HO-2). Heme 151-155 heme oxygenase 1 Homo sapiens 185-189 28298633-8 2017 Prior administration of hemin to renal IRI was associated with significant increase of the renal HO-1+ CD11b+ F4/80lo myeloid cells in comparison to control mice. Hemin 24-29 heme oxygenase 1 Homo sapiens 97-101 28109747-8 2017 Adaptation to acrolein is related to Nrf2 translocation, increased mRNA expression of gammaGCS, HO-1 and increased GSH levels and the increased GSH levels can explain the hormetic effect. Acrolein 14-22 heme oxygenase 1 Homo sapiens 96-100 28350337-7 2017 In addition, DBL enhanced cytoprotective effects through elevated antioxidant enzymes including heme oxygenase 1 (HO-1), catalase, glutathione peroxidase (GPx), and superoxide dismutase (SOD). disialyllactose 13-16 heme oxygenase 1 Homo sapiens 96-112 28350337-7 2017 In addition, DBL enhanced cytoprotective effects through elevated antioxidant enzymes including heme oxygenase 1 (HO-1), catalase, glutathione peroxidase (GPx), and superoxide dismutase (SOD). disialyllactose 13-16 heme oxygenase 1 Homo sapiens 114-118 28189672-0 2017 Valproic acid downregulates heme oxygenase-1 independently of Nrf2 by increasing ubiquitination and proteasomal degradation. Valproic Acid 0-13 heme oxygenase 1 Homo sapiens 28-44 27561292-7 2017 Meanwhile, allicin decreased NF-kappaB level and upregulated expression of proteins reduced by As2O3 including nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1, nicotinamide adenine dinucleotide phosphate:quinone oxidoreductase 1, and Kruppel-like factor 9 (KLF9). allicin 11-18 heme oxygenase 1 Homo sapiens 163-179 28107189-5 2017 Mechanistic studies demonstrated that DSF enhances DHA-induced cellular oxidative stress as evidenced by up-regulation of Nrf2-mediated heme oxygenase 1 (HO-1) gene transcription. Docosahexaenoic Acids 51-54 heme oxygenase 1 Homo sapiens 136-152 28107189-5 2017 Mechanistic studies demonstrated that DSF enhances DHA-induced cellular oxidative stress as evidenced by up-regulation of Nrf2-mediated heme oxygenase 1 (HO-1) gene transcription. Docosahexaenoic Acids 51-54 heme oxygenase 1 Homo sapiens 154-158 28055290-6 2017 In addition, we have demonstrated that heme degradation by HMOX1/HO-1 (heme oxygenase 1) is required and that Fe is essential for the formation of ALIS, as heme analogs lacking the central atom of Fe are not able to induce these structures. Heme 39-43 heme oxygenase 1 Homo sapiens 59-64 28055290-6 2017 In addition, we have demonstrated that heme degradation by HMOX1/HO-1 (heme oxygenase 1) is required and that Fe is essential for the formation of ALIS, as heme analogs lacking the central atom of Fe are not able to induce these structures. Heme 39-43 heme oxygenase 1 Homo sapiens 65-87 28055290-6 2017 In addition, we have demonstrated that heme degradation by HMOX1/HO-1 (heme oxygenase 1) is required and that Fe is essential for the formation of ALIS, as heme analogs lacking the central atom of Fe are not able to induce these structures. Heme 71-75 heme oxygenase 1 Homo sapiens 59-64 28082120-2 2017 Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Carbon Monoxide 145-160 heme oxygenase 1 Homo sapiens 30-34 28082120-2 2017 Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Iron 167-171 heme oxygenase 1 Homo sapiens 30-34 28082120-2 2017 Under several stress stimuli, HO-1 expression and activity is up-regulated to catalyze the rate-limiting enzymatic step of heme degradation into carbon monoxide, free iron, and biliverdin. Biliverdine 177-187 heme oxygenase 1 Homo sapiens 30-34 27297870-12 2017 Taken together, these results suggest that andrographolide suppresses hypoxia-induced pro-inflammatory ET-1 expression by activating Nrf2/HO-1, inhibiting p38 MAPK signaling, and promoting PHD2/3 expression. andrographolide 43-58 heme oxygenase 1 Homo sapiens 138-142 27297870-0 2017 Andrographolide inhibits hypoxia-induced HIF-1alpha-driven endothelin 1 secretion by activating Nrf2/HO-1 and promoting the expression of prolyl hydroxylases 2/3 in human endothelial cells. andrographolide 0-15 heme oxygenase 1 Homo sapiens 101-105 27297870-9 2017 Silencing Nrf2 and heme oxygenase 1 (HO-1) reversed the inhibitory effects of andrographolide on hypxoia-induced HIF-1alpha mRNA and protein expression. andrographolide 78-93 heme oxygenase 1 Homo sapiens 19-35 27996348-0 2017 Involvement of the activation of Nrf2/HO-1, p38 MAPK signaling pathways and endoplasmic reticulum stress in furazolidone induced cytotoxicity and S phase arrest in human hepatocyte L02 cells: modulation of curcumin. Furazolidone 108-120 heme oxygenase 1 Homo sapiens 38-42 27297870-9 2017 Silencing Nrf2 and heme oxygenase 1 (HO-1) reversed the inhibitory effects of andrographolide on hypxoia-induced HIF-1alpha mRNA and protein expression. andrographolide 78-93 heme oxygenase 1 Homo sapiens 37-41 26780453-0 2017 Protocatechualdehyde Protects Against Cerebral Ischemia-Reperfusion-Induced Oxidative Injury Via Protein Kinase Cepsilon/Nrf2/HO-1 Pathway. protocatechualdehyde 0-20 heme oxygenase 1 Homo sapiens 126-130 28115238-0 2017 NGF protects against oxygen and glucose deprivation-induced oxidative stress and apoptosis by up-regulation of HO-1 through MEK/ERK pathway. Oxygen 21-27 heme oxygenase 1 Homo sapiens 111-115 27996348-7 2017 Moreover, compared with the control, FZD exposure activated the protein and mRNA expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1), which were further activated by curcumin treatment. Curcumin 214-222 heme oxygenase 1 Homo sapiens 157-173 27996348-7 2017 Moreover, compared with the control, FZD exposure activated the protein and mRNA expression levels of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1), which were further activated by curcumin treatment. Curcumin 214-222 heme oxygenase 1 Homo sapiens 175-179 27996348-8 2017 These results reveal that curcumin could prevent FZD induced cytotoxicity and S phase arrest, which may involve the activation of Nrf2/HO-1 pathway and the inhibition of p38 MAPK pathway and ER stress. Curcumin 26-34 heme oxygenase 1 Homo sapiens 135-139 28088947-0 2017 Heme oxygenase-1 (HO-1)/carbon monoxide (CO) axis suppresses RANKL-induced osteoclastic differentiation by inhibiting redox-sensitive NF-kappaB activation. Carbon Monoxide 41-43 heme oxygenase 1 Homo sapiens 0-16 27996348-8 2017 These results reveal that curcumin could prevent FZD induced cytotoxicity and S phase arrest, which may involve the activation of Nrf2/HO-1 pathway and the inhibition of p38 MAPK pathway and ER stress. Furazolidone 49-52 heme oxygenase 1 Homo sapiens 135-139 28270832-0 2017 Actin Family Proteins in the Human INO80 Chromatin Remodeling Complex Exhibit Functional Roles in the Induction of Heme Oxygenase-1 with Hemin. Hemin 137-142 heme oxygenase 1 Homo sapiens 115-131 28108387-5 2017 Selegiline also reversed CSM-induced changes of anti-oxidant enzymes superoxide dismutase (SOD) and catalase (CAT) activities, GSH/GSSG ratio, as well expression of heme oxygenase-1 (HO-1) and NAD(P)H quinone dehydrogenase 1 (NQO1). Selegiline 0-10 heme oxygenase 1 Homo sapiens 165-181 28055957-5 2017 The latter is known to generate reactive oxygen species, and consequently, expression of the antioxidant enzyme heme oxygenase 1 (HMOX1) was significantly upregulated in 5-FU-treated cells, indicative for oxidative stress. Reactive Oxygen Species 32-55 heme oxygenase 1 Homo sapiens 112-128 28055957-5 2017 The latter is known to generate reactive oxygen species, and consequently, expression of the antioxidant enzyme heme oxygenase 1 (HMOX1) was significantly upregulated in 5-FU-treated cells, indicative for oxidative stress. Reactive Oxygen Species 32-55 heme oxygenase 1 Homo sapiens 130-135 28055957-5 2017 The latter is known to generate reactive oxygen species, and consequently, expression of the antioxidant enzyme heme oxygenase 1 (HMOX1) was significantly upregulated in 5-FU-treated cells, indicative for oxidative stress. Fluorouracil 170-174 heme oxygenase 1 Homo sapiens 112-128 28055957-5 2017 The latter is known to generate reactive oxygen species, and consequently, expression of the antioxidant enzyme heme oxygenase 1 (HMOX1) was significantly upregulated in 5-FU-treated cells, indicative for oxidative stress. Fluorouracil 170-174 heme oxygenase 1 Homo sapiens 130-135 28055957-7 2017 Also in vivo, 5-FU significantly induced hepatic ACOX1 and HMOX1 expression as well as JNK-activation, pro-inflammatory gene expression and immune cell infiltration. Fluorouracil 14-18 heme oxygenase 1 Homo sapiens 59-64 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Heme 32-36 heme oxygenase 1 Homo sapiens 16-20 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 16-20 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Carbon Monoxide 57-59 heme oxygenase 1 Homo sapiens 16-20 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Biliverdine 62-72 heme oxygenase 1 Homo sapiens 16-20 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Bilirubin 73-82 heme oxygenase 1 Homo sapiens 16-20 28088947-1 2017 Heme oxygenase (HO-1) catalyzes heme to carbon monoxide (CO), biliverdin/bilirubin, and iron and is known to prevent the pathogenesis of several human diseases. Iron 88-92 heme oxygenase 1 Homo sapiens 16-20 28119605-0 2016 Agrimonolide and Desmethylagrimonolide Induced HO-1 Expression in HepG2 Cells through Nrf2-Transduction and p38 Inactivation. Desmethylagrimonolide 17-38 heme oxygenase 1 Homo sapiens 47-51 28035393-9 2017 Fluoxetine inhibited MA-induced increases in the expression levels of serotonin transporter (SERT) and p-p38 mitogen-activated protein kinase (MAPK), and reversed the MA-induced decrease in nuclear Nrf2 and human heme oxygenase-1 in lungs. Fluoxetine 0-10 heme oxygenase 1 Homo sapiens 213-229 28120861-5 2017 CNT exposure activates an oxidative stress-dependent production of iron via Nrf2 nuclear translocation, Ferritin H and Heme oxygenase 1 translation. Iron 67-71 heme oxygenase 1 Homo sapiens 119-135 28116663-11 2017 HO-1 was inhibited by Sn-protoporphyrin, HO-1 inhibitor, and increased by cobalt protopophyrin, HO-1 inducer. tin protoporphyrin IX 22-39 heme oxygenase 1 Homo sapiens 0-4 28116663-11 2017 HO-1 was inhibited by Sn-protoporphyrin, HO-1 inhibitor, and increased by cobalt protopophyrin, HO-1 inducer. cobalt protopophyrin 74-94 heme oxygenase 1 Homo sapiens 0-4 28098837-0 2017 Casein Glycomacropeptide Hydrolysates Exert Cytoprotective Effect against Cellular Oxidative Stress by Up-Regulating HO-1 Expression in HepG2 Cells. glycomacropeptide hydrolysates 7-37 heme oxygenase 1 Homo sapiens 117-121 28098837-4 2017 Further, GHP concentration-dependently induced heme oxygenase-1 (HO-1) expression and increased nuclear factor-erythroid 2-related factor 2 (Nrf2) nuclear translocation. ghp 9-12 heme oxygenase 1 Homo sapiens 47-63 28098837-4 2017 Further, GHP concentration-dependently induced heme oxygenase-1 (HO-1) expression and increased nuclear factor-erythroid 2-related factor 2 (Nrf2) nuclear translocation. ghp 9-12 heme oxygenase 1 Homo sapiens 65-69 28098837-5 2017 Moreover, pretreatment of GHP increased the activation of p38 mitogen-activated protein kinase (p38 MAPK) and extracellular signal-regulated protein kinase 1/2 (ERK1/2), which were shown to contribute to Nrf2-mediated HO-1 expression. ghp 26-29 heme oxygenase 1 Homo sapiens 218-222 28161055-11 2017 In isolated primary trophoblasts, HSP70 and HO-1 were upregulated by increasing oxygen tension, but not by hyperglycemia or TNF-alpha. Oxygen 80-86 heme oxygenase 1 Homo sapiens 44-48 28119605-5 2016 Western blotting and luciferase assay revealed that exposure of HepG2 cells to agrimonolide or desmethylagrimonolide leads to increased heme oxygenase-1 (HO-1) expression by activating ARE through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). agrimonolide 79-91 heme oxygenase 1 Homo sapiens 136-152 28119605-5 2016 Western blotting and luciferase assay revealed that exposure of HepG2 cells to agrimonolide or desmethylagrimonolide leads to increased heme oxygenase-1 (HO-1) expression by activating ARE through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). agrimonolide 79-91 heme oxygenase 1 Homo sapiens 154-158 28119605-5 2016 Western blotting and luciferase assay revealed that exposure of HepG2 cells to agrimonolide or desmethylagrimonolide leads to increased heme oxygenase-1 (HO-1) expression by activating ARE through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). Desmethylagrimonolide 95-116 heme oxygenase 1 Homo sapiens 136-152 28119605-5 2016 Western blotting and luciferase assay revealed that exposure of HepG2 cells to agrimonolide or desmethylagrimonolide leads to increased heme oxygenase-1 (HO-1) expression by activating ARE through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). Desmethylagrimonolide 95-116 heme oxygenase 1 Homo sapiens 154-158 28119605-7 2016 In conclusion, our results indicated that transcriptional activation of Nrf2/ARE is critical in agrimonolide and desmethylagrimonolide-mediated HO-1 induction, which can be regulated partially by the blockade of p38 MAPK signaling pathway and inhibiting nuclear translocation of Nrf2. agrimonolide 96-108 heme oxygenase 1 Homo sapiens 144-148 28119605-7 2016 In conclusion, our results indicated that transcriptional activation of Nrf2/ARE is critical in agrimonolide and desmethylagrimonolide-mediated HO-1 induction, which can be regulated partially by the blockade of p38 MAPK signaling pathway and inhibiting nuclear translocation of Nrf2. Desmethylagrimonolide 113-134 heme oxygenase 1 Homo sapiens 144-148 27873232-7 2017 We demonstrated that heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway prevents placental stress and suppresses sFlt-1 and sEng release. Carbon Monoxide 45-60 heme oxygenase 1 Homo sapiens 21-37 27438141-0 2017 4-Hydroxyestradiol induces mammary epithelial cell transformation through Nrf2-mediated heme oxygenase-1 overexpression. 4-hydroxyestradiol 0-18 heme oxygenase 1 Homo sapiens 88-104 27438141-3 2017 In this study, 4-OHE2, but not E2, increased the expression of heme oxygenase-1 (HO-1), a sensor and regulator of oxidative stress, in MCF-10A cells. 4-hydroxyestradiol 15-21 heme oxygenase 1 Homo sapiens 63-79 27438141-3 2017 In this study, 4-OHE2, but not E2, increased the expression of heme oxygenase-1 (HO-1), a sensor and regulator of oxidative stress, in MCF-10A cells. 4-hydroxyestradiol 15-21 heme oxygenase 1 Homo sapiens 81-85 27438141-4 2017 Silencing the HO-1 gene in MCF-10A cells suppressed 4-OHE2-induced cell proliferation and transformation. 4-hydroxyestradiol 52-58 heme oxygenase 1 Homo sapiens 14-18 27438141-5 2017 In addition, subcutaneous administration of 4-OHE2 markedly enhanced the growth of the MDA-MB-231 human breast cancer xenografts, which was retarded by zinc protoporphyrin, a pharmacological inhibitor of HO-1. 4-hydroxyestradiol 44-50 heme oxygenase 1 Homo sapiens 204-208 27438141-5 2017 In addition, subcutaneous administration of 4-OHE2 markedly enhanced the growth of the MDA-MB-231 human breast cancer xenografts, which was retarded by zinc protoporphyrin, a pharmacological inhibitor of HO-1. zinc protoporphyrin 152-171 heme oxygenase 1 Homo sapiens 204-208 27438141-6 2017 4-OHE2-induced HO-1 expression was mediated by NF-E2-related factor 2 (Nrf2). 4-hydroxyestradiol 0-6 heme oxygenase 1 Homo sapiens 15-19 27873232-7 2017 We demonstrated that heme oxygenase-1 (HO-1)/carbon monoxide (CO) pathway prevents placental stress and suppresses sFlt-1 and sEng release. Carbon Monoxide 62-64 heme oxygenase 1 Homo sapiens 21-37 27943244-1 2017 OBJECTIVE: The present study was undertaken to investigate the genotype and allele frequencies of the variants in the four bilirubin metabolism genes (UGT1A1, OATP2, HMOX1, and BLVRA) and their association with hyperbilirubinemia. Bilirubin 123-132 heme oxygenase 1 Homo sapiens 166-171 28356018-0 2017 Enhancement of the Effect of Methyl Pyropheophorbide-a-Mediated Photodynamic Therapy was Achieved by Increasing ROS through Inhibition of Nrf2-HO-1 or Nrf2-ABCG2 Signaling. methyl pyropheophorbide-a 29-54 heme oxygenase 1 Homo sapiens 143-147 28356018-0 2017 Enhancement of the Effect of Methyl Pyropheophorbide-a-Mediated Photodynamic Therapy was Achieved by Increasing ROS through Inhibition of Nrf2-HO-1 or Nrf2-ABCG2 Signaling. Reactive Oxygen Species 112-115 heme oxygenase 1 Homo sapiens 143-147 28356018-7 2017 Nrf2 down -regulation increased reactive oxygen species (ROS) levels by attenuating antioxidants or pumping Mppa out of cells,which resulted from the inhibition of Nrf2-HO-1 or Nrf2- ABCG2 signaling. Reactive Oxygen Species 32-55 heme oxygenase 1 Homo sapiens 169-173 28503569-7 2017 These results indicate that TNFRI-Fc and hHO-1 overexpression may apparently induce free iron in the liver and exert oxidative stress by enhancing reactive oxygen species production and block normal postneonatal liver metabolism. Iron 89-93 heme oxygenase 1 Homo sapiens 41-46 28356018-7 2017 Nrf2 down -regulation increased reactive oxygen species (ROS) levels by attenuating antioxidants or pumping Mppa out of cells,which resulted from the inhibition of Nrf2-HO-1 or Nrf2- ABCG2 signaling. Reactive Oxygen Species 57-60 heme oxygenase 1 Homo sapiens 169-173 28107860-3 2017 RESULTS: Following exposure to the HO-1 inducer CoPPIX at all concentrations, the HBMEC cells apoptosis occurred, which could be prominently observed at 15 muM of 3 h exposure. coppix 48-54 heme oxygenase 1 Homo sapiens 35-39 28107860-6 2017 For the effect of the HO-1 inhibitor ZnPPIX, HBMEC cell morphology was mostly unchanged, but significant inhibitory effect on cell apoptosis was seen at 10 muM for the exposure period of 3 h (37.17% of baseline level). zinc protoporphyrin 37-43 heme oxygenase 1 Homo sapiens 22-26 27641735-10 2017 Restoration of HO-1 expression by hemin (20 mumol/L) abolished the inhibition of miR-1304 on cell growth and rescued miR-1304-induced apoptosis in A549 cells. Hemin 34-39 heme oxygenase 1 Homo sapiens 15-19 28503569-7 2017 These results indicate that TNFRI-Fc and hHO-1 overexpression may apparently induce free iron in the liver and exert oxidative stress by enhancing reactive oxygen species production and block normal postneonatal liver metabolism. Reactive Oxygen Species 147-170 heme oxygenase 1 Homo sapiens 41-46 27249374-12 2017 Sublethal concentration of acrolein upregulated HO-1 mRNA expression in retinal microvascular endothelial cells. Acrolein 27-35 heme oxygenase 1 Homo sapiens 48-52 28228072-8 2017 Dietary curcumin supplementation can also increase antioxidant activity through the induction of heme oxygenase-1, a scavenger of free radicals, and by reduction of reactive oxygen species and Nox-2. Curcumin 8-16 heme oxygenase 1 Homo sapiens 97-113 28382863-6 2017 Fenofibrate significantly upregulated heme oxygenase 1 (HO-1) at protein and mRNA levels in human glioblastoma LN-18 cells and rat brain astrocytes respectively, but nuclear factor kappalight- chain-enhancer of activated B cells (NFkappaB) was downregulated after fenofibrate treatment. Fenofibrate 0-11 heme oxygenase 1 Homo sapiens 38-54 28382863-6 2017 Fenofibrate significantly upregulated heme oxygenase 1 (HO-1) at protein and mRNA levels in human glioblastoma LN-18 cells and rat brain astrocytes respectively, but nuclear factor kappalight- chain-enhancer of activated B cells (NFkappaB) was downregulated after fenofibrate treatment. Fenofibrate 0-11 heme oxygenase 1 Homo sapiens 56-60 28382863-7 2017 Results showed that fenofibrate-induced upregulation of HO-1 expression were inhibited after LN-18 cells were transfected with 50nM small interfering RNA (siRNAs) for 48 hours to knockdown PPARalpha. Fenofibrate 20-31 heme oxygenase 1 Homo sapiens 56-60 28190391-0 2017 Novel Caffeic Acid Phenethyl Ester (Cape) Analogues as Inducers of Heme Oxygenase-1. caffeic acid phenethyl ester 6-34 heme oxygenase 1 Homo sapiens 67-83 28228072-8 2017 Dietary curcumin supplementation can also increase antioxidant activity through the induction of heme oxygenase-1, a scavenger of free radicals, and by reduction of reactive oxygen species and Nox-2. Free Radicals 130-143 heme oxygenase 1 Homo sapiens 97-113 28190391-0 2017 Novel Caffeic Acid Phenethyl Ester (Cape) Analogues as Inducers of Heme Oxygenase-1. caffeic acid phenethyl ester 36-40 heme oxygenase 1 Homo sapiens 67-83 28190391-4 2017 Caffeic acid phenethyl ester, a natural polyphenolic compound, behaves as HO-1 inducer and possesses a plethora of beneficial effects under oxidative stress conditions. caffeic acid phenethyl ester 0-28 heme oxygenase 1 Homo sapiens 74-78 28412905-2 2017 Among a battery of therapeutic targets, the heme oxygenase- 1 (HO-1)/carbon monoxide (CO) system has been evaluated for the development of new therapies against I/R injury. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 44-61 28190391-5 2017 OBJECTIVES: A small focused series of caffeic acid phenethyl ester (Cape) analogues was designed and synthesized with the aim of obtaining more potent HO-1 inducers. caffeic acid phenethyl ester 38-66 heme oxygenase 1 Homo sapiens 151-155 28190391-5 2017 OBJECTIVES: A small focused series of caffeic acid phenethyl ester (Cape) analogues was designed and synthesized with the aim of obtaining more potent HO-1 inducers. caffeic acid phenethyl ester 68-72 heme oxygenase 1 Homo sapiens 151-155 28190391-8 2017 VP961 tested to measure HO-1 protein expression and HO activity in in vitro system resulted more potent than the parent compound Cape both as inducer and as direct activator of the enzyme. caffeic acid phenethyl ester 129-133 heme oxygenase 1 Homo sapiens 24-28 28412905-3 2017 The enzyme HO-1 catalyzes the degradation of heme into three biologically active end products, namely biliverdin/bilirubin, CO and ferrous ion. Heme 45-49 heme oxygenase 1 Homo sapiens 11-15 28412905-3 2017 The enzyme HO-1 catalyzes the degradation of heme into three biologically active end products, namely biliverdin/bilirubin, CO and ferrous ion. Biliverdine 102-112 heme oxygenase 1 Homo sapiens 11-15 28412905-3 2017 The enzyme HO-1 catalyzes the degradation of heme into three biologically active end products, namely biliverdin/bilirubin, CO and ferrous ion. Bilirubin 113-122 heme oxygenase 1 Homo sapiens 11-15 28412905-10 2017 The regulation of HO-1/CO expression has been achieved either by genetic overexpression of HO-1 cDNA or pharmacological induction with drugs including curcumin and resveratrol. Curcumin 151-159 heme oxygenase 1 Homo sapiens 18-25 28412905-10 2017 The regulation of HO-1/CO expression has been achieved either by genetic overexpression of HO-1 cDNA or pharmacological induction with drugs including curcumin and resveratrol. Curcumin 151-159 heme oxygenase 1 Homo sapiens 18-22 28412905-10 2017 The regulation of HO-1/CO expression has been achieved either by genetic overexpression of HO-1 cDNA or pharmacological induction with drugs including curcumin and resveratrol. Resveratrol 164-175 heme oxygenase 1 Homo sapiens 18-25 28412905-10 2017 The regulation of HO-1/CO expression has been achieved either by genetic overexpression of HO-1 cDNA or pharmacological induction with drugs including curcumin and resveratrol. Resveratrol 164-175 heme oxygenase 1 Homo sapiens 18-22 27908781-3 2017 To elucidate the mechanisms involved, here we assess whether the HO-1 downstream metabolites biliverdin (BV) and/or iron mediate the HO-1 antiviral effect. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 65-69 27867098-3 2017 Exogenously administered ammonia, given as ammonium chloride or ammonium hydroxide, or endogenously generated ammonia stimulated HO-1 protein expression in cultured human and murine endothelial cells. Ammonia 25-32 heme oxygenase 1 Homo sapiens 129-133 27908781-3 2017 To elucidate the mechanisms involved, here we assess whether the HO-1 downstream metabolites biliverdin (BV) and/or iron mediate the HO-1 antiviral effect. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 133-137 27867098-3 2017 Exogenously administered ammonia, given as ammonium chloride or ammonium hydroxide, or endogenously generated ammonia stimulated HO-1 protein expression in cultured human and murine endothelial cells. Ammonium Hydroxide 64-82 heme oxygenase 1 Homo sapiens 129-133 27867098-3 2017 Exogenously administered ammonia, given as ammonium chloride or ammonium hydroxide, or endogenously generated ammonia stimulated HO-1 protein expression in cultured human and murine endothelial cells. Ammonia 110-117 heme oxygenase 1 Homo sapiens 129-133 27908781-3 2017 To elucidate the mechanisms involved, here we assess whether the HO-1 downstream metabolites biliverdin (BV) and/or iron mediate the HO-1 antiviral effect. Biliverdine 105-107 heme oxygenase 1 Homo sapiens 65-69 27908781-3 2017 To elucidate the mechanisms involved, here we assess whether the HO-1 downstream metabolites biliverdin (BV) and/or iron mediate the HO-1 antiviral effect. Biliverdine 105-107 heme oxygenase 1 Homo sapiens 133-137 27908781-3 2017 To elucidate the mechanisms involved, here we assess whether the HO-1 downstream metabolites biliverdin (BV) and/or iron mediate the HO-1 antiviral effect. Iron 116-120 heme oxygenase 1 Homo sapiens 133-137 27908781-8 2017 Collectively, our findings identify a HO-1-BV/BR-NO-cGMP/PKG cascade as a novel pathway underlying the host cell antiviral effect. Cyclic GMP 52-56 heme oxygenase 1 Homo sapiens 38-42 27922667-6 2017 Moreover, while downregulation of HO-1 by the small-molecule inhibitor tin protoporphyrin (SnPP) promoted migration, upregulation of HO-1 by the small-molecule activator cobalt protoporphyrin (CoPP) showed the opposite effect. tin protoporphyrin IX 71-89 heme oxygenase 1 Homo sapiens 34-38 27976481-5 2017 Accumulation of ROS and subsequent activation of NRF2, p53, AP-1 and NF-kappaB-dependent pathways, with downstream activation of antioxidant mechanisms (e.g., SOD2 and HMOX1 expression), is observed in the UV-treated cells. Reactive Oxygen Species 16-19 heme oxygenase 1 Homo sapiens 168-173 27922667-6 2017 Moreover, while downregulation of HO-1 by the small-molecule inhibitor tin protoporphyrin (SnPP) promoted migration, upregulation of HO-1 by the small-molecule activator cobalt protoporphyrin (CoPP) showed the opposite effect. S-Nitroso-N-propionyl-D,L-penicillamine 91-95 heme oxygenase 1 Homo sapiens 34-38 27922667-6 2017 Moreover, while downregulation of HO-1 by the small-molecule inhibitor tin protoporphyrin (SnPP) promoted migration, upregulation of HO-1 by the small-molecule activator cobalt protoporphyrin (CoPP) showed the opposite effect. cobaltiprotoporphyrin 170-191 heme oxygenase 1 Homo sapiens 133-137 27720465-10 2017 CONCLUSION: Cinnamaldehyde has two independent biological activities; namely, an inhibitory action on AHR activation and an antioxidant effect mediated by NRF2/HO1 signaling. cinnamaldehyde 12-26 heme oxygenase 1 Homo sapiens 160-163 27856716-1 2017 Endogenous carbon monoxide (CO) levels are recognized as a surrogate marker for activity of heme oxygenase-1, which is induced by various factors, including hypoxia and oxidative stress. Carbon Monoxide 11-26 heme oxygenase 1 Homo sapiens 92-108 27856716-1 2017 Endogenous carbon monoxide (CO) levels are recognized as a surrogate marker for activity of heme oxygenase-1, which is induced by various factors, including hypoxia and oxidative stress. Carbon Monoxide 28-30 heme oxygenase 1 Homo sapiens 92-108 27720465-8 2017 Cinnamaldehyde also activated the NRF2/HO1 pathway and significantly alleviated the production of reactive oxygen species in keratinocytes. cinnamaldehyde 0-14 heme oxygenase 1 Homo sapiens 39-42 27959441-0 2017 Protective effects of Cambodian medicinal plants on tert-butyl hydroperoxide-induced hepatotoxicity via Nrf2-mediated heme oxygenase-1. tert-Butylhydroperoxide 52-76 heme oxygenase 1 Homo sapiens 118-134 28740855-6 2017 In addition, fidarestat augmented the HG-induced Nrf2 expression and activity and also upregulated the expression of Nrf2-dependent proteins such as hemeoxygenase-1 (HO1) and NQO1 in Thp1 cells. fidarestat 13-23 heme oxygenase 1 Homo sapiens 149-169 27959441-9 2017 These results indicated that, of the 64 Cambodian plants, P. weberi and T. crispa exhibited hepatoprotective effects on t-BHP-induced cytotoxicity in HepG2 cells, possibly by the induction of Nrf2-mediated expression of HO-1. tert-Butylhydroperoxide 120-125 heme oxygenase 1 Homo sapiens 220-224 28123555-3 2017 HO-1 expression was evaluated using immunohistochemistry in 147 formalin-fixed tissue specimens. Formaldehyde 64-72 heme oxygenase 1 Homo sapiens 0-4 26742524-0 2017 High-Glucose-Derived Oxidative Stress-Dependent Heme Oxygenase-1 Expression from Astrocytes Contributes to the Neuronal Apoptosis. high-glucose 0-12 heme oxygenase 1 Homo sapiens 48-64 26742524-6 2017 We demonstrated that HG induced HO-1 expression via a reactive oxygen species (ROS)-dependent signaling pathway. Reactive Oxygen Species 54-77 heme oxygenase 1 Homo sapiens 32-36 26742524-6 2017 We demonstrated that HG induced HO-1 expression via a reactive oxygen species (ROS)-dependent signaling pathway. Reactive Oxygen Species 79-82 heme oxygenase 1 Homo sapiens 32-36 26742524-9 2017 These results indicated that in brain astrocytes, activation of MAPK-mediated NF-kappaB and c-Fos/AP-1 cascades by Nox/ROS and mitoROS-dependent events is essential for HO-1 up-regulation induced by HG. Reactive Oxygen Species 119-122 heme oxygenase 1 Homo sapiens 169-173 26742524-9 2017 These results indicated that in brain astrocytes, activation of MAPK-mediated NF-kappaB and c-Fos/AP-1 cascades by Nox/ROS and mitoROS-dependent events is essential for HO-1 up-regulation induced by HG. mitoros 127-134 heme oxygenase 1 Homo sapiens 169-173 26993631-3 2017 In addition we tested also the hypothesis that CO can decrease the pre-existing condition of oxidative stress in the mouse model for the human medical condition systemic lupus erythematosus by increasing two protective enzymes heme-oxygenase-1 (HO-1), and superoxide dismutase-2 (SOD-2). Carbon Monoxide 47-49 heme oxygenase 1 Homo sapiens 227-243 26993631-3 2017 In addition we tested also the hypothesis that CO can decrease the pre-existing condition of oxidative stress in the mouse model for the human medical condition systemic lupus erythematosus by increasing two protective enzymes heme-oxygenase-1 (HO-1), and superoxide dismutase-2 (SOD-2). Carbon Monoxide 47-49 heme oxygenase 1 Homo sapiens 245-249 28694915-6 2017 Overall, our study is the first to demonstrate that the cytoprotective actions of halophenols involve their antiapoptotic, antioxidant, and anti-inflammatory abilities, which are mediated by the upregulation of Nrf2-dependent HO-1 expression and reductions in ROS and TNF-alpha generation via the activation of Erk1/2 and PI3K/Akt in EA.hy926 cells. halophenols 82-93 heme oxygenase 1 Homo sapiens 226-230 28694915-0 2017 Heme Oxygenase-1, a Key Enzyme for the Cytoprotective Actions of Halophenols by Upregulating Nrf2 Expression via Activating Erk1/2 and PI3K/Akt in EA.hy926 Cells. halophenols 65-76 heme oxygenase 1 Homo sapiens 0-16 28473878-0 2017 HO-1 Is Essential for Tetrahydroxystilbene Glucoside Mediated Mitochondrial Biogenesis and Anti-Inflammation Process in LPS-Treated RAW264.7 Macrophages. 2',3',4',5'-tetrahydroxystilbene-2-O-beta-D-glucoside 22-52 heme oxygenase 1 Homo sapiens 0-4 28473878-7 2017 Zinc Protoporphyrin (ZnPP), a selective inhibitor of HO-1 activity, was able to attenuate TSG mediated mitochondrial biogenesis and anti-inflammatory process. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 53-57 28473878-7 2017 Zinc Protoporphyrin (ZnPP), a selective inhibitor of HO-1 activity, was able to attenuate TSG mediated mitochondrial biogenesis and anti-inflammatory process. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 53-57 28694915-6 2017 Overall, our study is the first to demonstrate that the cytoprotective actions of halophenols involve their antiapoptotic, antioxidant, and anti-inflammatory abilities, which are mediated by the upregulation of Nrf2-dependent HO-1 expression and reductions in ROS and TNF-alpha generation via the activation of Erk1/2 and PI3K/Akt in EA.hy926 cells. ros 260-263 heme oxygenase 1 Homo sapiens 226-230 28694915-3 2017 In the present study, we demonstrated that these halophenols exhibited significant protective effects against H2O2-induced injury in EA.hy926 cells by inhibition of apoptosis and ROS and TNF-alpha production, as well as induction of the upregulation of HO-1, the magnitude of which correlated with their cytoprotective actions. halophenols 49-60 heme oxygenase 1 Homo sapiens 253-257 28694915-3 2017 In the present study, we demonstrated that these halophenols exhibited significant protective effects against H2O2-induced injury in EA.hy926 cells by inhibition of apoptosis and ROS and TNF-alpha production, as well as induction of the upregulation of HO-1, the magnitude of which correlated with their cytoprotective actions. Hydrogen Peroxide 110-114 heme oxygenase 1 Homo sapiens 253-257 28694915-7 2017 HO-1 may thus be an important potential target for further research into the cytoprotective actions of halophenols. halophenols 103-114 heme oxygenase 1 Homo sapiens 0-4 28694916-0 2017 The Protective Effect of Indole-3-Acetic Acid (IAA) on H2O2-Damaged Human Dental Pulp Stem Cells Is Mediated by the AKT Pathway and Involves Increased Expression of the Transcription Factor Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2) and Its Downstream Target Heme Oxygenase 1 (HO-1). indoleacetic acid 25-45 heme oxygenase 1 Homo sapiens 267-283 28694916-0 2017 The Protective Effect of Indole-3-Acetic Acid (IAA) on H2O2-Damaged Human Dental Pulp Stem Cells Is Mediated by the AKT Pathway and Involves Increased Expression of the Transcription Factor Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2) and Its Downstream Target Heme Oxygenase 1 (HO-1). indoleacetic acid 25-45 heme oxygenase 1 Homo sapiens 285-289 28694916-0 2017 The Protective Effect of Indole-3-Acetic Acid (IAA) on H2O2-Damaged Human Dental Pulp Stem Cells Is Mediated by the AKT Pathway and Involves Increased Expression of the Transcription Factor Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2) and Its Downstream Target Heme Oxygenase 1 (HO-1). indoleacetic acid 47-50 heme oxygenase 1 Homo sapiens 267-283 28694916-0 2017 The Protective Effect of Indole-3-Acetic Acid (IAA) on H2O2-Damaged Human Dental Pulp Stem Cells Is Mediated by the AKT Pathway and Involves Increased Expression of the Transcription Factor Nuclear Factor-Erythroid 2-Related Factor 2 (Nrf2) and Its Downstream Target Heme Oxygenase 1 (HO-1). indoleacetic acid 47-50 heme oxygenase 1 Homo sapiens 285-289 28694916-8 2017 To further investigate the mechanism of IAA, Nrf2-related antioxidant pathway was examined and the results showed that the level of Nrf2 and HO-1 expressions, stimulated by H2O2, decreased after treatment of IAA. Hydrogen Peroxide 173-177 heme oxygenase 1 Homo sapiens 141-145 28694916-8 2017 To further investigate the mechanism of IAA, Nrf2-related antioxidant pathway was examined and the results showed that the level of Nrf2 and HO-1 expressions, stimulated by H2O2, decreased after treatment of IAA. indoleacetic acid 40-43 heme oxygenase 1 Homo sapiens 141-145 28694916-9 2017 Moreover, IAA treatment protected hDPSCs against H2O2-induced oxidative stress via increased expression of Nrf2 and HO-1, mediated by the AKT pathway. Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 116-120 27627465-5 2017 RESULTS: We found that salidroside pretreatment upregulated Nrf2 translocation to the nucleus and transcription activity in HaCaT cells, as reflected by the increased nuclear accumulation of Nrf2 as well as the gene and protein expression of downstream Nrf2 antioxidants, including NQO1 and HO-1. rhodioloside 23-34 heme oxygenase 1 Homo sapiens 291-295 29158871-3 2017 Because a crosstalk exists between nitric oxide (NO) and HO-1 in promotion of cell survival under oxidative stress, we designed novel NO-releasing molecules also capable to induce HO-1. Nitric Oxide 35-47 heme oxygenase 1 Homo sapiens 180-184 29158871-4 2017 Starting from curcumin and caffeic acid phenethyl ester (CAPE), two known HO-1 inducers, the molecules were chemically modified by acylation with 4-bromo-butanoyl chloride and 2-chloro-propanoyl chloride, respectively, and then treated in the dark with AgNO3 to obtain the nitrate derivatives VP10/12 and VP10/39. Curcumin 14-22 heme oxygenase 1 Homo sapiens 74-78 29158871-4 2017 Starting from curcumin and caffeic acid phenethyl ester (CAPE), two known HO-1 inducers, the molecules were chemically modified by acylation with 4-bromo-butanoyl chloride and 2-chloro-propanoyl chloride, respectively, and then treated in the dark with AgNO3 to obtain the nitrate derivatives VP10/12 and VP10/39. caffeic acid phenethyl ester 27-55 heme oxygenase 1 Homo sapiens 74-78 29158871-4 2017 Starting from curcumin and caffeic acid phenethyl ester (CAPE), two known HO-1 inducers, the molecules were chemically modified by acylation with 4-bromo-butanoyl chloride and 2-chloro-propanoyl chloride, respectively, and then treated in the dark with AgNO3 to obtain the nitrate derivatives VP10/12 and VP10/39. caffeic acid phenethyl ester 57-61 heme oxygenase 1 Homo sapiens 74-78 30263473-0 2016 Effect of (-)-epigallocatechin-3-gallate on anti-inflammatory response via heme oxygenase-1 induction during adipocyte-macrophage interactions. epigallocatechin gallate 10-40 heme oxygenase 1 Homo sapiens 75-91 28255307-6 2017 Then, MSC or MSC-HO-1 was cocultured with retinal ganglion cells (RGC-5) in H2O2-simulated oxidative condition and their protection on RGC-5 was systemically valuated in vitro. Hydrogen Peroxide 76-80 heme oxygenase 1 Homo sapiens 17-21 28255307-7 2017 Compared with MSC, MSC-HO-1 significantly attenuated H2O2-induced injury of RGC-5, including decrease in cellular ROS level and apoptosis, activation of antiapoptotic proteins p-Akt and Bcl-2, and blockage of proapoptotic proteins cleaved caspase 3 and Bax. Hydrogen Peroxide 53-57 heme oxygenase 1 Homo sapiens 23-27 28255307-7 2017 Compared with MSC, MSC-HO-1 significantly attenuated H2O2-induced injury of RGC-5, including decrease in cellular ROS level and apoptosis, activation of antiapoptotic proteins p-Akt and Bcl-2, and blockage of proapoptotic proteins cleaved caspase 3 and Bax. ros 114-117 heme oxygenase 1 Homo sapiens 23-27 28751936-7 2017 Furthermore, we also found that the protective effects of MaR 1 on lung tissue injury and oxidative stress were reversed by HO-1 activity inhibitor, Znpp-IX. zinc protoporphyrin 149-156 heme oxygenase 1 Homo sapiens 124-128 28751936-8 2017 Nrf-2 transcription factor inhibitor, brusatol, significantly decreased MaR 1-induced nuclear Nrf-2 and cytosolic HO-1 expression. brusatol 38-46 heme oxygenase 1 Homo sapiens 114-118 30263473-1 2016 In this study, we examined the effects of (-)-epigallocatechin-3-gallate (EGCG) on anti-inflammatory responses through the induction of heme oxygenase-1 (HO-1) in cocultured macrophages and adipocytes. epigallocatechin gallate 42-72 heme oxygenase 1 Homo sapiens 136-152 30263473-1 2016 In this study, we examined the effects of (-)-epigallocatechin-3-gallate (EGCG) on anti-inflammatory responses through the induction of heme oxygenase-1 (HO-1) in cocultured macrophages and adipocytes. epigallocatechin gallate 42-72 heme oxygenase 1 Homo sapiens 154-158 30263473-1 2016 In this study, we examined the effects of (-)-epigallocatechin-3-gallate (EGCG) on anti-inflammatory responses through the induction of heme oxygenase-1 (HO-1) in cocultured macrophages and adipocytes. epigallocatechin gallate 74-78 heme oxygenase 1 Homo sapiens 136-152 30263473-1 2016 In this study, we examined the effects of (-)-epigallocatechin-3-gallate (EGCG) on anti-inflammatory responses through the induction of heme oxygenase-1 (HO-1) in cocultured macrophages and adipocytes. epigallocatechin gallate 74-78 heme oxygenase 1 Homo sapiens 154-158 30263473-6 2016 These results indicate that EGCG inhibited inflammatory responses by suppressing the production of proinflammatory cytokines through HO-1 induction during adipocyte-macrophage interaction. epigallocatechin gallate 28-32 heme oxygenase 1 Homo sapiens 133-137 28050122-14 2016 CDECM induced nuclear factor erythroid-2 related factor 2 (Nrf2) mediated-antioxidant enzyme heme oxygenase-1 (HO-1). cdecm 0-5 heme oxygenase 1 Homo sapiens 93-109 27795439-3 2017 In this study, inhibition of PRRSV replication was demonstrated to be mediated by carbon monoxide (CO), a downstream metabolite of HO-1. Carbon Monoxide 82-97 heme oxygenase 1 Homo sapiens 131-135 27997582-2 2016 The cytoprotective enzyme heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation and it has been shown to exert anti-inflammatory functions. Heme 26-30 heme oxygenase 1 Homo sapiens 44-48 27795439-3 2017 In this study, inhibition of PRRSV replication was demonstrated to be mediated by carbon monoxide (CO), a downstream metabolite of HO-1. Carbon Monoxide 99-101 heme oxygenase 1 Homo sapiens 131-135 27795439-12 2017 Carbon monoxide (CO), a metabolite of HO-1, has been shown to have antimicrobial and antiviral activities in infected cells. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 38-42 27795439-12 2017 Carbon monoxide (CO), a metabolite of HO-1, has been shown to have antimicrobial and antiviral activities in infected cells. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 38-42 27863411-6 2016 Meanwhile, sesamin was found to up-regulate the expression of Nrf2 and HO-1. sesamin 11-18 heme oxygenase 1 Homo sapiens 71-75 27912883-7 2016 RESULTS: Miltirone-induced Nrf2 and HO-1 expression was related to mitogen-activated protein kinase (MAPK) pathways. miltirone 9-18 heme oxygenase 1 Homo sapiens 36-40 27912883-9 2016 However, miltirone-induced Nrf2/HO-1 expression can only be effectively blocked by JNK inhibitor SP600125. miltirone 9-18 heme oxygenase 1 Homo sapiens 32-36 27912883-9 2016 However, miltirone-induced Nrf2/HO-1 expression can only be effectively blocked by JNK inhibitor SP600125. pyrazolanthrone 97-105 heme oxygenase 1 Homo sapiens 32-36 27912883-10 2016 CONCLUSION: Our findings reveal that miltirone exerts protective functions on endothelial cells in response to ox-LDL-induced oxidative stress, and does so via Nrf2/HO-1, which provides novel insights into the antioxidant capacity of miltirone. miltirone 37-46 heme oxygenase 1 Homo sapiens 165-169 27912883-0 2016 Miltirone protects human EA.hy926 endothelial cells from oxidized low-density lipoprotein-derived oxidative stress via a heme oxygenase-1 and MAPK/Nrf2 dependent pathway. miltirone 0-9 heme oxygenase 1 Homo sapiens 121-137 27912883-4 2016 STUDY DESIGN/METHOD: In the present study, miltirone increased the expression of nuclear translocation and transcriptional activities of NF-E2-related factor 2 (Nrf2), which led to augmented expression of antioxidant-response element (ARE)-dependent heme oxygenase-1 (HO-1) and NAD(P)H-quinone oxidoreductase 1 (NQO1). miltirone 43-52 heme oxygenase 1 Homo sapiens 250-266 27912883-5 2016 Inhibition of Nrf2/HO-1 by RNA interference abolished miltirone-induced cytoprotective effects against ox-LDL, which suggested that Nrf2 and the downstream expression of HO-1 are required for the functional effects of miltirone. miltirone 54-63 heme oxygenase 1 Homo sapiens 19-23 27912883-5 2016 Inhibition of Nrf2/HO-1 by RNA interference abolished miltirone-induced cytoprotective effects against ox-LDL, which suggested that Nrf2 and the downstream expression of HO-1 are required for the functional effects of miltirone. miltirone 54-63 heme oxygenase 1 Homo sapiens 170-174 27912883-5 2016 Inhibition of Nrf2/HO-1 by RNA interference abolished miltirone-induced cytoprotective effects against ox-LDL, which suggested that Nrf2 and the downstream expression of HO-1 are required for the functional effects of miltirone. miltirone 218-227 heme oxygenase 1 Homo sapiens 19-23 27912883-5 2016 Inhibition of Nrf2/HO-1 by RNA interference abolished miltirone-induced cytoprotective effects against ox-LDL, which suggested that Nrf2 and the downstream expression of HO-1 are required for the functional effects of miltirone. miltirone 218-227 heme oxygenase 1 Homo sapiens 170-174 27829220-0 2016 Gene expression profile induced by arsenic trioxide in chronic lymphocytic leukemia cells reveals a central role for heme oxygenase-1 in apoptosis and regulation of matrix metalloproteinase-9. Arsenic Trioxide 35-51 heme oxygenase 1 Homo sapiens 117-133 27829220-4 2016 ATO modulated many genes, largely involved in oxidative stress, being HMOX1 the most upregulated gene, also induced at the protein level. Arsenic Trioxide 0-3 heme oxygenase 1 Homo sapiens 70-75 27829220-9 2016 Cell viability analyses upon modulation of HMOX1 expression or activity demonstrated that HMOX1 had a pro-apoptotic role and enhanced the cytotoxic effect of ATO in CLL cells. Arsenic Trioxide 158-161 heme oxygenase 1 Homo sapiens 43-48 27829220-9 2016 Cell viability analyses upon modulation of HMOX1 expression or activity demonstrated that HMOX1 had a pro-apoptotic role and enhanced the cytotoxic effect of ATO in CLL cells. Arsenic Trioxide 158-161 heme oxygenase 1 Homo sapiens 90-95 27725188-0 2016 Omeprazole induces heme oxygenase-1 in fetal human pulmonary microvascular endothelial cells via hydrogen peroxide-independent Nrf2 signaling pathway. Omeprazole 0-10 heme oxygenase 1 Homo sapiens 19-35 30461238-0 2016 Translational toxicology from basic sciences to clinical toxicology Toxicological significance of heme oxygenase-1 induction, the enzyme of carbon monoxide production. Carbon Monoxide 140-155 heme oxygenase 1 Homo sapiens 98-114 26593436-7 2016 IsoLQ also induced the expression of Nrf2 target phase II enzymes, such as heme oxygenase-1, glutamate-cysteine ligase catalytic subunit and NAD(P)H:quinone oxidoreductase 1. isolq 0-5 heme oxygenase 1 Homo sapiens 75-91 27931585-8 2016 Further, resveratrol activated the expression of sirtuin-1 and Nrf2, and inhibited the expression of osteopontin, runt-related transcription factor 2, and heme oxygenase-1. Resveratrol 9-20 heme oxygenase 1 Homo sapiens 155-171 27853236-7 2016 Luteolin enhanced antioxidant defense system based on Keap1, Nrf2, HO-1, NQO1, and KLF9. Luteolin 0-8 heme oxygenase 1 Homo sapiens 67-71 27725188-11 2016 Although N-acetyl cysteine (NAC) significantly decreased H2O2 levels in OM-treated cells, we observed that OM further increased HO-1 mRNA and protein expression in NAC-pretreated compared to vehicle-pretreated cells, suggesting that OM induces HO-1 via H2O2-independent mechanisms. Acetylcysteine 164-167 heme oxygenase 1 Homo sapiens 128-132 27725188-11 2016 Although N-acetyl cysteine (NAC) significantly decreased H2O2 levels in OM-treated cells, we observed that OM further increased HO-1 mRNA and protein expression in NAC-pretreated compared to vehicle-pretreated cells, suggesting that OM induces HO-1 via H2O2-independent mechanisms. Acetylcysteine 164-167 heme oxygenase 1 Homo sapiens 244-248 27725188-12 2016 In conclusion, we provide evidence that OM transcriptionally induces HO-1 via AhR - and H2O2 - independent, but Nrf2 - dependent mechanisms. Hydrogen Peroxide 88-92 heme oxygenase 1 Homo sapiens 69-73 27725188-0 2016 Omeprazole induces heme oxygenase-1 in fetal human pulmonary microvascular endothelial cells via hydrogen peroxide-independent Nrf2 signaling pathway. Hydrogen Peroxide 97-114 heme oxygenase 1 Homo sapiens 19-35 27725188-9 2016 Interestingly, the concentration at which OM induced HO-1 also increased H2O2 levels. Hydrogen Peroxide 73-77 heme oxygenase 1 Homo sapiens 53-57 27725188-10 2016 Furthermore, H2O2 independently augmented HO-1 expression. Hydrogen Peroxide 13-17 heme oxygenase 1 Homo sapiens 42-46 27591243-13 2016 14,15-EET treatment resulted in a significant reduction in IL-6, IL-8, and MCP-1 secretion, and increased accumulation of Nrf2 and expression of HO-1. 14,15-epoxy-5,8,11-eicosatrienoic acid 0-9 heme oxygenase 1 Homo sapiens 145-149 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. Metals 223-228 heme oxygenase 1 Homo sapiens 57-73 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. Metals 223-228 heme oxygenase 1 Homo sapiens 75-80 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. Metals 291-296 heme oxygenase 1 Homo sapiens 57-73 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. Metals 291-296 heme oxygenase 1 Homo sapiens 75-80 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. ros 301-304 heme oxygenase 1 Homo sapiens 57-73 27782264-7 2016 Phase 2 enzymes in term of antioxidant response, such as heme oxygenase 1 (HMOX1) and the regulating subunit of the glutamate-cysteine ligase (GCLM) were slightly upregulated, but these observations may be linked solely to metal homeostasis disruptions, as these actors are involved in both metal and ROS responses. ros 301-304 heme oxygenase 1 Homo sapiens 75-80 27412172-10 2016 Moreover, MG also protects adipocytes from oxidative stress by alleviating intracellular reactive oxygen species and activating Nrf2, HO-1, and PRDX3. methyl gallate 10-12 heme oxygenase 1 Homo sapiens 134-138 27374075-9 2016 Multiple logistic regression analysis, however, showed that only rs2364723 significantly reduced levels of serum HMOX1 in T2DM patients for the GG genotype carriers compared with participants with CG+CC genotype. cysteinylglycine 197-199 heme oxygenase 1 Homo sapiens 113-118 27694475-8 2016 Finally, peripheral blood monocytes (PBMCs) from elderly patients with P-NTM also demonstrated attenuated HO-1 responses after M. avium stimulation and increased cell death due to cellular necrosis (9.69% +- 2.02) compared with apoptosis (4.75% +- 0.98). p-ntm 71-76 heme oxygenase 1 Homo sapiens 106-110 27418236-0 2016 Andrographolide protects liver cells from H2O2 induced cell death by upregulation of Nrf-2/HO-1 mediated via adenosine A2a receptor signalling. andrographolide 0-15 heme oxygenase 1 Homo sapiens 91-95 27418236-0 2016 Andrographolide protects liver cells from H2O2 induced cell death by upregulation of Nrf-2/HO-1 mediated via adenosine A2a receptor signalling. Hydrogen Peroxide 42-46 heme oxygenase 1 Homo sapiens 91-95 27418236-6 2016 RESULTS: We clearly show that andrographolide via adenosine A2A receptor signalling leads to activation of p38 MAP kinase, resulting in upregulation of Nrf-2, its translocation to nucleus and activation of HO-1. andrographolide 30-45 heme oxygenase 1 Homo sapiens 206-210 27418236-9 2016 CONCLUSIONS: Thus, andrographolide probably by binding to adenosine A2a receptor activates Nrf-2 transcription and also inhibits its exclusion from the nucleus by inactivating GSK-3beta, together resulting in activation of HO-1. andrographolide 19-34 heme oxygenase 1 Homo sapiens 223-227 27671470-8 2016 A mechanistic study showed that Co-Cr alloys activates the NRF2 pathway and up-regulate antioxidant enzymes including heme oxygenase-1 (HO-1). co-cr 32-37 heme oxygenase 1 Homo sapiens 118-134 27505325-0 2016 GT-repeat length polymorphism in heme oxygenase-1 promoter determines the effect of cilostazol on vascular smooth muscle cells. Cilostazol 84-94 heme oxygenase 1 Homo sapiens 33-49 27505325-2 2016 The aim of this study is to investigate whether the inhibitory effect of cilostazol on VSMC proliferation is operated via heme oxygenase-1 (HO-1). Cilostazol 73-83 heme oxygenase 1 Homo sapiens 122-138 27505325-2 2016 The aim of this study is to investigate whether the inhibitory effect of cilostazol on VSMC proliferation is operated via heme oxygenase-1 (HO-1). Cilostazol 73-83 heme oxygenase 1 Homo sapiens 140-144 27505325-2 2016 The aim of this study is to investigate whether the inhibitory effect of cilostazol on VSMC proliferation is operated via heme oxygenase-1 (HO-1). vsmc 87-91 heme oxygenase 1 Homo sapiens 122-138 27505325-2 2016 The aim of this study is to investigate whether the inhibitory effect of cilostazol on VSMC proliferation is operated via heme oxygenase-1 (HO-1). vsmc 87-91 heme oxygenase 1 Homo sapiens 140-144 27505325-4 2016 Treatment of human VSMCs with cilostazol enhanced the expression of HO-1, which was mainly regulated at the transcriptional level. Cilostazol 30-40 heme oxygenase 1 Homo sapiens 68-72 27505325-5 2016 Small interfering RNA knock-down of HO-1 attenuated the inhibitory effect of cilostazol on VSMC proliferation, suggesting the critical role of HO-1 in cilostazol effect. Cilostazol 77-87 heme oxygenase 1 Homo sapiens 36-40 27505325-5 2016 Small interfering RNA knock-down of HO-1 attenuated the inhibitory effect of cilostazol on VSMC proliferation, suggesting the critical role of HO-1 in cilostazol effect. Cilostazol 77-87 heme oxygenase 1 Homo sapiens 143-147 27505325-5 2016 Small interfering RNA knock-down of HO-1 attenuated the inhibitory effect of cilostazol on VSMC proliferation, suggesting the critical role of HO-1 in cilostazol effect. vsmc 91-95 heme oxygenase 1 Homo sapiens 36-40 27505325-5 2016 Small interfering RNA knock-down of HO-1 attenuated the inhibitory effect of cilostazol on VSMC proliferation, suggesting the critical role of HO-1 in cilostazol effect. Cilostazol 151-161 heme oxygenase 1 Homo sapiens 36-40 27505325-5 2016 Small interfering RNA knock-down of HO-1 attenuated the inhibitory effect of cilostazol on VSMC proliferation, suggesting the critical role of HO-1 in cilostazol effect. Cilostazol 151-161 heme oxygenase 1 Homo sapiens 143-147 27505325-6 2016 The transcriptional responsiveness of HO-1 to cilostazol was inversely correlated with the length of GT-repeat in human HO-1 promoter. Cilostazol 46-56 heme oxygenase 1 Homo sapiens 38-42 27505325-6 2016 The transcriptional responsiveness of HO-1 to cilostazol was inversely correlated with the length of GT-repeat in human HO-1 promoter. Cilostazol 46-56 heme oxygenase 1 Homo sapiens 120-124 27505325-7 2016 Deletion and mutational analysis of HO-1 promoter along with chromatin immunoprecipitation showed that cyclic AMP response element (CRE)-binding protein (CREB) participated in cilostazol-induced HO-1 transcription. Cyclic AMP 103-113 heme oxygenase 1 Homo sapiens 36-40 27505325-7 2016 Deletion and mutational analysis of HO-1 promoter along with chromatin immunoprecipitation showed that cyclic AMP response element (CRE)-binding protein (CREB) participated in cilostazol-induced HO-1 transcription. Cyclic AMP 103-113 heme oxygenase 1 Homo sapiens 195-199 27505325-7 2016 Deletion and mutational analysis of HO-1 promoter along with chromatin immunoprecipitation showed that cyclic AMP response element (CRE)-binding protein (CREB) participated in cilostazol-induced HO-1 transcription. Cilostazol 176-186 heme oxygenase 1 Homo sapiens 36-40 27505325-7 2016 Deletion and mutational analysis of HO-1 promoter along with chromatin immunoprecipitation showed that cyclic AMP response element (CRE)-binding protein (CREB) participated in cilostazol-induced HO-1 transcription. Cilostazol 176-186 heme oxygenase 1 Homo sapiens 195-199 27505325-8 2016 Furthermore, cilostazol triggered a linkage between the CRE and GT-repeat regions in the HO-1 promoter. Cilostazol 13-23 heme oxygenase 1 Homo sapiens 89-93 27505325-9 2016 The promoting effect of cilostazol on HO-1 expression, proliferation inhibition, and chromatin conformation in the HO-1 promoter was greater in VSMCs from subjects with shorter GT-repeat alleles than those with longer alleles. Cilostazol 24-34 heme oxygenase 1 Homo sapiens 38-42 27505325-9 2016 The promoting effect of cilostazol on HO-1 expression, proliferation inhibition, and chromatin conformation in the HO-1 promoter was greater in VSMCs from subjects with shorter GT-repeat alleles than those with longer alleles. Cilostazol 24-34 heme oxygenase 1 Homo sapiens 115-119 27505325-10 2016 CONCLUSIONS: Cilostazol inhibits VSMC proliferation involving an association between CREB and HO-1. Cilostazol 13-23 heme oxygenase 1 Homo sapiens 94-98 27505325-10 2016 CONCLUSIONS: Cilostazol inhibits VSMC proliferation involving an association between CREB and HO-1. vsmc 33-37 heme oxygenase 1 Homo sapiens 94-98 27505325-11 2016 The length polymorphism of GT-repeat in human HO-1 promoter determines the effect of cilostazol. Cilostazol 85-95 heme oxygenase 1 Homo sapiens 46-50 27397680-7 2016 Alternatively, GGT5 overexpression induces heme oxygenase 1 (HO-1) expression, which, as a key catalyst responsible for the oxidative degradation of heme, may inhibit the activities of the cytochrome P450 monooxygenases, thus substantially impairing testicular steroidogenesis. Heme 43-47 heme oxygenase 1 Homo sapiens 61-65 27397680-8 2016 These results, coupled with the differential roles of mitogen-activated protein kinases and HO-1 signaling in spermatogenesis, lead us to propose a model in which a delicate balance between these two pathways modulated by the GGT5/oxidative stress cascade plays a central role during LH-stimulated steroidogenesis. Luteinizing Hormone 284-286 heme oxygenase 1 Homo sapiens 92-96 27662012-0 2016 Mutating heme oxygenase-1 into a peroxidase causes a defect in bilirubin synthesis associated with microcytic anemia and severe hyperinflammation. Bilirubin 63-72 heme oxygenase 1 Homo sapiens 9-25 27257045-1 2016 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme, which may be involved in the pathogenesis of AKI. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 27257045-1 2016 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme, which may be involved in the pathogenesis of AKI. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 27257045-2 2016 Length polymorphisms in the number of GT dinucleotide repeats in the HO-1 gene (HMOX1) promoter inversely associate with HMOX1 mRNA expression. gt dinucleotide 38-53 heme oxygenase 1 Homo sapiens 69-73 27257045-2 2016 Length polymorphisms in the number of GT dinucleotide repeats in the HO-1 gene (HMOX1) promoter inversely associate with HMOX1 mRNA expression. gt dinucleotide 38-53 heme oxygenase 1 Homo sapiens 80-85 27257045-2 2016 Length polymorphisms in the number of GT dinucleotide repeats in the HO-1 gene (HMOX1) promoter inversely associate with HMOX1 mRNA expression. gt dinucleotide 38-53 heme oxygenase 1 Homo sapiens 121-126 27571925-5 2016 Iron, one of HO-1 catalytic products, was an important mediator in this regulation. Iron 0-4 heme oxygenase 1 Homo sapiens 13-17 27283929-4 2016 AIMS: To compare effects of hemin and placebo infusions on HO1 activity and protein, GE, autonomic function, and gastrointestinal symptoms in diabetic gastroparesis. Hemin 28-33 heme oxygenase 1 Homo sapiens 59-62 27283929-12 2016 Further studies are necessary to ascertain whether more frequent hemin infusions or other drugs would have a more sustained effect on HO1 and improve GE. Hemin 65-70 heme oxygenase 1 Homo sapiens 134-137 27678042-0 2016 Calycosin suppresses expression of pro-inflammatory cytokines via the activation of p62/Nrf2-linked heme oxygenase 1 in rheumatoid arthritis synovial fibroblasts. 7,3'-dihydroxy-4'-methoxyisoflavone 0-9 heme oxygenase 1 Homo sapiens 100-116 27620489-8 2016 Cd exposure to p53-overexpressing YD10B cells enhanced Cd-induced HO-1 and LC3-II levels, whereas genetic knockdown of p53 in YD8 cells resulted in the suppression of Cd-induced levels of HO-1 and LC3-II, indicating that p53 is required in the sensing of HO-1 and induction of autophagy. Cadmium 55-57 heme oxygenase 1 Homo sapiens 66-70 27795400-1 2016 Heme oxygenase-1 (HO-1) is a stress response antioxidant enzyme which catalyzes the degradation of heme released during inflammation. Heme 99-103 heme oxygenase 1 Homo sapiens 18-22 27795400-4 2016 To address this controversy, we administered tin protoporphyrin IX (SnPPIX), a well-characterized HO-1 enzymatic inhibitor, to mice during acute M. tuberculosis infection. tin protoporphyrin IX 45-66 heme oxygenase 1 Homo sapiens 98-102 27795400-4 2016 To address this controversy, we administered tin protoporphyrin IX (SnPPIX), a well-characterized HO-1 enzymatic inhibitor, to mice during acute M. tuberculosis infection. tin protoporphyrin IX 68-74 heme oxygenase 1 Homo sapiens 98-102 27795400-13 2016 Here, we describe a potential strategy for treating TB based on pharmacological inhibition of the host heme-degrading enzyme HO-1. Heme 103-107 heme oxygenase 1 Homo sapiens 125-129 27562971-8 2016 Decreasing NADH pharmacologically with MTOB or genetically blocking CtBP1 with siRNA upregulated the cyclin-dependent genes (p15 and p21) and proapoptotic regulators (NOXA and PERP), attenuated proliferation, corrected the glycolytic reprogramming phenotype of PH-Fibs, and augmented transcription of the anti-inflammatory gene HMOX1. NAD 11-15 heme oxygenase 1 Homo sapiens 328-333 27620489-0 2016 Heme oxygenase-1-mediated apoptosis under cadmium-induced oxidative stress is regulated by autophagy, which is sensitized by tumor suppressor p53. Cadmium 42-49 heme oxygenase 1 Homo sapiens 0-16 27411481-6 2016 HO-1 expression levels were determined with western blot in monocytes before and after induction of HO-1 with cobalt protoporphyrin (CoPP) with or without CXCL4. cobaltiprotoporphyrin 110-131 heme oxygenase 1 Homo sapiens 100-104 27411481-8 2016 RESULTS: SSc patients have lower plasma levels of bilirubin, suggestive of an aberrant HO-1 function. Bilirubin 50-59 heme oxygenase 1 Homo sapiens 87-91 27411481-9 2016 We demonstrated low HO-1 expression in immune cells from SSc patients, whereas induction with CoPP was able to restore HO-1 levels in DCs from SSc patients, almost normalizing the increased TLR response observed in SSc. cobaltiprotoporphyrin 94-98 heme oxygenase 1 Homo sapiens 119-123 27411481-10 2016 Co-exposure to CXCL4 completely abrogated CoPP-induced HO-1 expression, suggesting that the high CXCL4 levels present in SSc patients block the normal induction of HO-1 and its function. cobaltiprotoporphyrin 42-46 heme oxygenase 1 Homo sapiens 55-59 27411481-10 2016 Co-exposure to CXCL4 completely abrogated CoPP-induced HO-1 expression, suggesting that the high CXCL4 levels present in SSc patients block the normal induction of HO-1 and its function. cobaltiprotoporphyrin 42-46 heme oxygenase 1 Homo sapiens 164-168 27620489-4 2016 Here we show that the role of HO-1 under Cd-induced oxidative stress is dependent upon autophagy, which is sensitized by the tumor suppressor p53. Cadmium 41-43 heme oxygenase 1 Homo sapiens 30-34 27620489-7 2016 In both cell lines, Cd exposure resulted in caspase-3-mediated PARP-1 cleavage and the induction of CHOP, LC3-II, and HO-1, which were limited in YD10B and H1299 cells exposed to high concentrations of Cd. Cadmium 20-22 heme oxygenase 1 Homo sapiens 106-122 27620489-9 2016 The inhibition of autophagy using small interfering RNA (siRNA) for the autophagy-related gene atg5 enhanced HO-1, CHOP, and PARP-1 cleavage induced by Cd. Cadmium 152-154 heme oxygenase 1 Homo sapiens 109-113 27620489-7 2016 In both cell lines, Cd exposure resulted in caspase-3-mediated PARP-1 cleavage and the induction of CHOP, LC3-II, and HO-1, which were limited in YD10B and H1299 cells exposed to high concentrations of Cd. Cadmium 202-204 heme oxygenase 1 Homo sapiens 106-122 27620489-10 2016 However, transfection with HO-1 siRNA increased Cd-induced LC3-II, and suppressed the expression of CHOP and cleavage of PARP-1. Cadmium 48-50 heme oxygenase 1 Homo sapiens 27-31 27620489-8 2016 Cd exposure to p53-overexpressing YD10B cells enhanced Cd-induced HO-1 and LC3-II levels, whereas genetic knockdown of p53 in YD8 cells resulted in the suppression of Cd-induced levels of HO-1 and LC3-II, indicating that p53 is required in the sensing of HO-1 and induction of autophagy. Cadmium 0-2 heme oxygenase 1 Homo sapiens 66-70 27620489-8 2016 Cd exposure to p53-overexpressing YD10B cells enhanced Cd-induced HO-1 and LC3-II levels, whereas genetic knockdown of p53 in YD8 cells resulted in the suppression of Cd-induced levels of HO-1 and LC3-II, indicating that p53 is required in the sensing of HO-1 and induction of autophagy. Cadmium 0-2 heme oxygenase 1 Homo sapiens 188-192 27598034-0 2016 15-Deoxy-Delta12,14-prostaglandin J2 stabilizes hypoxia inducible factor-1alpha through induction of heme oxygenase-1 and direct modification ofprolyl-4-hydroxylase 2. 14-prostaglandin j2 17-36 heme oxygenase 1 Homo sapiens 101-117 27620489-8 2016 Cd exposure to p53-overexpressing YD10B cells enhanced Cd-induced HO-1 and LC3-II levels, whereas genetic knockdown of p53 in YD8 cells resulted in the suppression of Cd-induced levels of HO-1 and LC3-II, indicating that p53 is required in the sensing of HO-1 and induction of autophagy. Cadmium 0-2 heme oxygenase 1 Homo sapiens 188-192 27620489-8 2016 Cd exposure to p53-overexpressing YD10B cells enhanced Cd-induced HO-1 and LC3-II levels, whereas genetic knockdown of p53 in YD8 cells resulted in the suppression of Cd-induced levels of HO-1 and LC3-II, indicating that p53 is required in the sensing of HO-1 and induction of autophagy. Cadmium 55-57 heme oxygenase 1 Homo sapiens 66-70 27375080-0 2016 Propyl gallate sensitizes human lung cancer cells to cisplatin-induced apoptosis by targeting heme oxygenase-1 for TRC8-mediated degradation. Propyl Gallate 0-14 heme oxygenase 1 Homo sapiens 94-110 27375080-0 2016 Propyl gallate sensitizes human lung cancer cells to cisplatin-induced apoptosis by targeting heme oxygenase-1 for TRC8-mediated degradation. Cisplatin 53-62 heme oxygenase 1 Homo sapiens 94-110 27375080-3 2016 In the present study, we investigated whether PG exhibit an anti-cancer effect through modulating HO-1 activation. Propyl Gallate 46-48 heme oxygenase 1 Homo sapiens 98-102 27375080-4 2016 In human non-small cell lung cancer (NSCLC) cells, treatment with PG dose-dependently diminished HO-1 protein levels without changing its mRNA levels and consequently decreased HO-1 activity. Propyl Gallate 66-68 heme oxygenase 1 Homo sapiens 97-101 27375080-4 2016 In human non-small cell lung cancer (NSCLC) cells, treatment with PG dose-dependently diminished HO-1 protein levels without changing its mRNA levels and consequently decreased HO-1 activity. Propyl Gallate 66-68 heme oxygenase 1 Homo sapiens 177-181 27375080-5 2016 PG also significantly enhanced the sensitivity of NSCLC cells to cisplatin-induced apoptosis, and this effect was attenuated by overexpression of HO-1. Propyl Gallate 0-2 heme oxygenase 1 Homo sapiens 146-150 27375080-5 2016 PG also significantly enhanced the sensitivity of NSCLC cells to cisplatin-induced apoptosis, and this effect was attenuated by overexpression of HO-1. Cisplatin 65-74 heme oxygenase 1 Homo sapiens 146-150 27375080-6 2016 Mechanistically, PG exerted its chemosensitization effect by down-regulating HO-1 protein expression through a TRC8 (translocation in renal carcinoma, chromosome 8)-mediated ubiquitin-proteasome pathway. Propyl Gallate 17-19 heme oxygenase 1 Homo sapiens 77-81 27375080-7 2016 Collectively, our data provide the potential application of PG in combination chemotherapy to enhance drug sensitivity in lung cancer by targeting HO-1. Propyl Gallate 60-62 heme oxygenase 1 Homo sapiens 147-151 27822409-13 2016 NAC (ROS scavenging agent) could inhibit the migration and invasion through reversing EMT and decrease the expression of HO-1. Reactive Oxygen Species 18-21 heme oxygenase 1 Homo sapiens 134-138 27822409-14 2016 What"s more, Znpp (HO-1 inhibitor) impaired the migration and invasion through reversing EMT. zinc protoporphyrin 13-17 heme oxygenase 1 Homo sapiens 19-23 27822409-15 2016 In conclusion, our results suggest that autophagy inhibition may promote EMT through ROS/HO-1 pathway in ovarian cancer cells. Reactive Oxygen Species 85-88 heme oxygenase 1 Homo sapiens 89-93 27698370-4 2016 Both inhibition of Na/K-ATPase oxidant amplification with pNaKtide and induction of heme oxygenase-1 (HO-1) with cobalt protoporphyrin (CoPP) markedly attenuated the development of phenotypical features of uremic cardiomyopathy. cobaltiprotoporphyrin 113-134 heme oxygenase 1 Homo sapiens 84-100 27698370-4 2016 Both inhibition of Na/K-ATPase oxidant amplification with pNaKtide and induction of heme oxygenase-1 (HO-1) with cobalt protoporphyrin (CoPP) markedly attenuated the development of phenotypical features of uremic cardiomyopathy. cobaltiprotoporphyrin 136-140 heme oxygenase 1 Homo sapiens 84-100 27488535-5 2016 Inhibition of HO-1 by tin protoporphyrin (SnPP) or siRNA downregulated Pax3/7-FoxO1 target genes and induced a myogenic program in RMS. tin protoporphyrin IX 22-40 heme oxygenase 1 Homo sapiens 14-18 27488535-5 2016 Inhibition of HO-1 by tin protoporphyrin (SnPP) or siRNA downregulated Pax3/7-FoxO1 target genes and induced a myogenic program in RMS. S-Nitroso-N-propionyl-D,L-penicillamine 42-46 heme oxygenase 1 Homo sapiens 14-18 27488535-6 2016 These effects were not mediated by altered myoD expression; instead, cells with elevated HO-1 produced less reactive oxygen species, resulting in nuclear localization of HDAC4 and miR-206 repression. Reactive Oxygen Species 108-131 heme oxygenase 1 Homo sapiens 89-93 27598034-4 2016 Pretreatment with zinc protoporphyrin IX, a pharmacological inhibitor of heme oxygenase-1 (HO-1), as well as siRNA knockdown of HO-1 gene in MCF-7 cells attenuated 15d-PGJ2-mediated HIF-1alpha accumulation. zinc protoporphyrin 18-40 heme oxygenase 1 Homo sapiens 73-89 27598034-4 2016 Pretreatment with zinc protoporphyrin IX, a pharmacological inhibitor of heme oxygenase-1 (HO-1), as well as siRNA knockdown of HO-1 gene in MCF-7 cells attenuated 15d-PGJ2-mediated HIF-1alpha accumulation. 15-deoxy-delta(12,14)-prostaglandin J2 164-172 heme oxygenase 1 Homo sapiens 91-95 27598034-4 2016 Pretreatment with zinc protoporphyrin IX, a pharmacological inhibitor of heme oxygenase-1 (HO-1), as well as siRNA knockdown of HO-1 gene in MCF-7 cells attenuated 15d-PGJ2-mediated HIF-1alpha accumulation. 15-deoxy-delta(12,14)-prostaglandin J2 164-172 heme oxygenase 1 Homo sapiens 128-132 27598034-5 2016 15d-PGJ2 treatment increased intracellular production of reactive oxygen species (ROS), which was mediated by HO-1 induction. 15-deoxy-delta(12,14)-prostaglandin J2 0-8 heme oxygenase 1 Homo sapiens 110-114 27598034-5 2016 15d-PGJ2 treatment increased intracellular production of reactive oxygen species (ROS), which was mediated by HO-1 induction. Reactive Oxygen Species 57-80 heme oxygenase 1 Homo sapiens 110-114 27598034-5 2016 15d-PGJ2 treatment increased intracellular production of reactive oxygen species (ROS), which was mediated by HO-1 induction. Reactive Oxygen Species 82-85 heme oxygenase 1 Homo sapiens 110-114 27598034-6 2016 Preincubation of MCF-7 cells with trolox, a water-soluble form of vitamin E, attenuated 15d-PGJ2-induced HIF-1alpha expression although HO-1 expression was unchanged. 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid 34-40 heme oxygenase 1 Homo sapiens 136-140 27598034-7 2016 This finding suggests that ROS accumulated as a consequence of HO-1 up-regulation can enhance HIF-1alpha expression in MCF-7 cells treated with 15d-PGJ2. Reactive Oxygen Species 27-30 heme oxygenase 1 Homo sapiens 63-67 27598034-7 2016 This finding suggests that ROS accumulated as a consequence of HO-1 up-regulation can enhance HIF-1alpha expression in MCF-7 cells treated with 15d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 144-152 heme oxygenase 1 Homo sapiens 63-67 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Heme 96-100 heme oxygenase 1 Homo sapiens 46-51 27573874-0 2016 Resveratrol pretreatment attenuates injury and promotes proliferation of neural stem cells following oxygen-glucose deprivation/reoxygenation by upregulating the expression of Nrf2, HO-1 and NQO1 in vitro. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 182-186 27573874-10 2016 These findings suggested that resveratrol attenuated injury and promoted proliferation of the NSCs, at least in part, by upregulating the expression of Nrf2, HO-1 and NQO1 following OGD/R injury in vitro. Resveratrol 30-41 heme oxygenase 1 Homo sapiens 158-162 27428762-5 2016 On the other hand, Mycophenolate mofetil induced GPX1 mRNA, although it suppressed mRNA level of UGT1A4/1A9/2B7/2B10, GSTA1/O1/T1, GSR and HMOX1 in HepG2 cells. Mycophenolic Acid 19-40 heme oxygenase 1 Homo sapiens 139-144 27222475-1 2016 The nuclear factor erythroid-derived two-like 2-antioxidant response element (Nrf2-ARE) pathway and its downstream antioxidant enzyme heme oxygenase-1 (HMOX1 or HO-1) play essential roles in H2 O2 -induced oxidative damage in human melanocytes. Hydrogen Peroxide 191-196 heme oxygenase 1 Homo sapiens 134-150 27222475-1 2016 The nuclear factor erythroid-derived two-like 2-antioxidant response element (Nrf2-ARE) pathway and its downstream antioxidant enzyme heme oxygenase-1 (HMOX1 or HO-1) play essential roles in H2 O2 -induced oxidative damage in human melanocytes. Hydrogen Peroxide 191-196 heme oxygenase 1 Homo sapiens 152-157 27222475-1 2016 The nuclear factor erythroid-derived two-like 2-antioxidant response element (Nrf2-ARE) pathway and its downstream antioxidant enzyme heme oxygenase-1 (HMOX1 or HO-1) play essential roles in H2 O2 -induced oxidative damage in human melanocytes. Hydrogen Peroxide 191-196 heme oxygenase 1 Homo sapiens 161-165 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Carbon Monoxide 104-119 heme oxygenase 1 Homo sapiens 46-51 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Carbon Monoxide 121-123 heme oxygenase 1 Homo sapiens 46-51 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Iron 126-138 heme oxygenase 1 Homo sapiens 46-51 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 46-51 27454768-6 2016 Furthermore, quercetin induced activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) and subsequent mRNA and protein expression of the antioxidant enzymes, heme oxygenase-1 (HO-1), NAD(P)H dehydrogenase, quinone 1, and glutamate-cysteine ligase. Quercetin 13-22 heme oxygenase 1 Homo sapiens 171-187 27604527-1 2016 Heme oxygenases are composed of two isozymes, Hmox1 and Hmox2, that catalyze the degradation of heme to carbon monoxide (CO), ferrous iron, and biliverdin, the latter of which is subsequently converted to bilirubin. Bilirubin 205-214 heme oxygenase 1 Homo sapiens 46-51 27685634-13 2016 The functional link between HO-1 activity and bile acid toxicity was established in RIP3-deficient primary hepatocytes. Bile Acids and Salts 46-55 heme oxygenase 1 Homo sapiens 28-32 27538638-0 2016 Inhibition of beta-amyloid-induced neurotoxicity by pinocembrin through Nrf2/HO-1 pathway in SH-SY5Y cells. pinocembrin 52-63 heme oxygenase 1 Homo sapiens 77-81 27542238-5 2016 In addition, treatment with ANM eliminated HG-induced reactive oxygen species (ROS) through the induction of anti-oxidant genes, HO-1 and NQO-1 via transcriptional activation of Nrf2. Reactive Oxygen Species 79-82 heme oxygenase 1 Homo sapiens 129-133 27609268-0 2016 Depression-like behaviors and heme oxygenase-1 are regulated by Lycopene in lipopolysaccharide-induced neuroinflammation. Lycopene 64-72 heme oxygenase 1 Homo sapiens 30-46 27538638-8 2016 PCB treatment also resulted in an increase in Nrf2 protein levels and subsequent induction of heme oxygenase-1(HO-1) expression in SH-SY5Y cells. pinocembrin 0-3 heme oxygenase 1 Homo sapiens 94-115 27609268-10 2016 Furthermore, lycopene decreased LPS-induced expression of IL-1beta and HO-1 in the hippocampus together with decreasing level of IL-6 and TNF-alpha in the plasma. Lycopene 13-21 heme oxygenase 1 Homo sapiens 71-75 27609268-11 2016 Taken together, these results suggest that lycopene can attenuate LPS-induced inflammation and depression-like behaviors, which may be involved in regulating HO-1 in the hippocampus. Lycopene 43-51 heme oxygenase 1 Homo sapiens 158-162 27538638-9 2016 RNA interference-mediated knockdown of Nrf2 expression suppressed the PCB-induced HO-1 expression. pinocembrin 70-73 heme oxygenase 1 Homo sapiens 82-86 27538638-10 2016 Notably, we found that the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) markedly diminished the neuroprotective effect of PCB against Abeta-mediated neurotoxicity. zinc protoporphyrin 42-64 heme oxygenase 1 Homo sapiens 27-31 27538638-10 2016 Notably, we found that the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) markedly diminished the neuroprotective effect of PCB against Abeta-mediated neurotoxicity. zinc protoporphyrin 66-70 heme oxygenase 1 Homo sapiens 27-31 27538638-10 2016 Notably, we found that the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) markedly diminished the neuroprotective effect of PCB against Abeta-mediated neurotoxicity. pinocembrin 122-125 heme oxygenase 1 Homo sapiens 27-31 27538638-11 2016 Taken together, these results indicated that PCB protects SH-SY5Y cells from Abeta25-35-induced neurotoxicity through activation of Nrf2/HO-1 pathways. pinocembrin 45-48 heme oxygenase 1 Homo sapiens 137-141 26404762-7 2016 TMP inhibited arsenic-induced activations of JNK, p38 MAPK, ERK, AP-1 and Nrf2 and block HO-1 protein expression. Arsenic 14-21 heme oxygenase 1 Homo sapiens 89-93 26404762-11 2016 In summary, our study highlighted a role of HO-1 in the protection against arsenic-induced cytotoxicity downstream from the primary targets of TMP and further indicated that TMP may be used as a potential therapeutic agent in the treatment of arsenic-induced nephrotoxicity. Arsenic 75-82 heme oxygenase 1 Homo sapiens 44-48 26404762-0 2016 2,3,5,6-Tetramethylpyrazine (TMP) down-regulated arsenic-induced heme oxygenase-1 and ARS2 expression by inhibiting Nrf2, NF-kappaB, AP-1 and MAPK pathways in human proximal tubular cells. tetramethylpyrazine 0-27 heme oxygenase 1 Homo sapiens 65-81 26404762-11 2016 In summary, our study highlighted a role of HO-1 in the protection against arsenic-induced cytotoxicity downstream from the primary targets of TMP and further indicated that TMP may be used as a potential therapeutic agent in the treatment of arsenic-induced nephrotoxicity. tetramethylpyrazine 174-177 heme oxygenase 1 Homo sapiens 44-48 26404762-0 2016 2,3,5,6-Tetramethylpyrazine (TMP) down-regulated arsenic-induced heme oxygenase-1 and ARS2 expression by inhibiting Nrf2, NF-kappaB, AP-1 and MAPK pathways in human proximal tubular cells. tetramethylpyrazine 29-32 heme oxygenase 1 Homo sapiens 65-81 26404762-0 2016 2,3,5,6-Tetramethylpyrazine (TMP) down-regulated arsenic-induced heme oxygenase-1 and ARS2 expression by inhibiting Nrf2, NF-kappaB, AP-1 and MAPK pathways in human proximal tubular cells. Arsenic 49-56 heme oxygenase 1 Homo sapiens 65-81 26404762-11 2016 In summary, our study highlighted a role of HO-1 in the protection against arsenic-induced cytotoxicity downstream from the primary targets of TMP and further indicated that TMP may be used as a potential therapeutic agent in the treatment of arsenic-induced nephrotoxicity. Arsenic 243-250 heme oxygenase 1 Homo sapiens 44-48 26404762-2 2016 The present study demonstrated that arsenic exposure resulted in protein and enzymatic induction of heme oxygenase-1 (HO-1) in dose- and time-dependent manners in HK-2 cells. Arsenic 36-43 heme oxygenase 1 Homo sapiens 100-116 26938875-2 2016 Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). Carbon Monoxide 135-150 heme oxygenase 1 Homo sapiens 0-16 26404762-2 2016 The present study demonstrated that arsenic exposure resulted in protein and enzymatic induction of heme oxygenase-1 (HO-1) in dose- and time-dependent manners in HK-2 cells. Arsenic 36-43 heme oxygenase 1 Homo sapiens 118-122 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. zinc protoporphyrin 36-55 heme oxygenase 1 Homo sapiens 9-13 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. zinc protoporphyrin 57-61 heme oxygenase 1 Homo sapiens 9-13 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. zinc protoporphyrin 57-61 heme oxygenase 1 Homo sapiens 177-181 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. Arsenic 73-80 heme oxygenase 1 Homo sapiens 9-13 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. Arsenic 73-80 heme oxygenase 1 Homo sapiens 177-181 26404762-3 2016 Blocking HO-1 enzymatic activity by zinc protoporphyrin (ZnPP) augmented arsenic-induced apoptosis, ROS production and mitochondrial dysfunction, suggesting a critical role for HO-1 as a renal protectant in this procession. ros 100-103 heme oxygenase 1 Homo sapiens 9-13 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. tetramethylpyrazine 19-22 heme oxygenase 1 Homo sapiens 36-40 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. tetramethylpyrazine 19-22 heme oxygenase 1 Homo sapiens 101-105 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. Arsenic 52-59 heme oxygenase 1 Homo sapiens 36-40 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. ros 68-71 heme oxygenase 1 Homo sapiens 36-40 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. ros 87-90 heme oxygenase 1 Homo sapiens 36-40 26404762-4 2016 On the other hand, TMP, upstream of HO-1, inhibited arsenic-induced ROS production and ROS-dependent HO-1 expression. ros 87-90 heme oxygenase 1 Homo sapiens 101-105 26404762-6 2016 Our results revealed that the regulation of arsenic-induced HO-1 expression was performed through multiple ROS-dependent signal pathways and the corresponding transcription factors, including p38 MAPK and JNK (but not ERK), AP-1, Nrf2 and NF-kappaB. Arsenic 44-51 heme oxygenase 1 Homo sapiens 60-64 26404762-6 2016 Our results revealed that the regulation of arsenic-induced HO-1 expression was performed through multiple ROS-dependent signal pathways and the corresponding transcription factors, including p38 MAPK and JNK (but not ERK), AP-1, Nrf2 and NF-kappaB. ros 107-110 heme oxygenase 1 Homo sapiens 60-64 26404762-7 2016 TMP inhibited arsenic-induced activations of JNK, p38 MAPK, ERK, AP-1 and Nrf2 and block HO-1 protein expression. tetramethylpyrazine 0-3 heme oxygenase 1 Homo sapiens 89-93 27375190-0 2016 Curcumin attenuates quinocetone induced apoptosis and inflammation via the opposite modulation of Nrf2/HO-1 and NF-kB pathway in human hepatocyte L02 cells. Curcumin 0-8 heme oxygenase 1 Homo sapiens 103-107 27375190-6 2016 Meanwhile, curcumin pretreatment markedly down-regulated the expression of nuclear factor -kB (NF-kB) and iNOS mRNAs, but up-regulated the expressions of Nrf2 and HO-1 mRNAs, compared to the QCT alone group. Curcumin 11-19 heme oxygenase 1 Homo sapiens 163-167 27375190-7 2016 Zinc protoporphyrin IX, a HO-1 inhibitor, markedly partly abolished the cytoprotective effect of curcumin against QCT-induced caspase activation, NF-kB mRNA expression. zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 26-30 27375190-7 2016 Zinc protoporphyrin IX, a HO-1 inhibitor, markedly partly abolished the cytoprotective effect of curcumin against QCT-induced caspase activation, NF-kB mRNA expression. Curcumin 97-105 heme oxygenase 1 Homo sapiens 26-30 27373419-7 2016 However, resveratrol prevented these effects, protecting hippocampal astrocytes against azide-induced cytotoxicity through the heme-oxygenase-1 (HO-1) pathway and inhibiting p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor kappa B (NFkappaB) activation. Resveratrol 9-20 heme oxygenase 1 Homo sapiens 127-143 27373419-7 2016 However, resveratrol prevented these effects, protecting hippocampal astrocytes against azide-induced cytotoxicity through the heme-oxygenase-1 (HO-1) pathway and inhibiting p38 mitogen-activated protein kinase (p38 MAPK) and nuclear factor kappa B (NFkappaB) activation. Azides 88-93 heme oxygenase 1 Homo sapiens 127-143 27146412-0 2016 Resveratrol Protects Oxidative Stress-Induced Intestinal Epithelial Barrier Dysfunction by Upregulating Heme Oxygenase-1 Expression. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 104-120 27146412-9 2016 Moreover, Res which protected Caco-2 cells from H2O2-induced oxidative damage clearly reduced malondialdehyde level and intracellular reactive oxygen species accumulation, but increased the expression levels of superoxide dismutase and heme oxygenase-1 (HO-1). Hydrogen Peroxide 48-52 heme oxygenase 1 Homo sapiens 236-252 27146412-9 2016 Moreover, Res which protected Caco-2 cells from H2O2-induced oxidative damage clearly reduced malondialdehyde level and intracellular reactive oxygen species accumulation, but increased the expression levels of superoxide dismutase and heme oxygenase-1 (HO-1). Hydrogen Peroxide 48-52 heme oxygenase 1 Homo sapiens 254-258 27146412-11 2016 Interestingly, these effects of Res were abolished by the HO-1 inhibitor zinc protoporphyrin or knockdown of HO-1 by siRNA. zinc protoporphyrin 73-92 heme oxygenase 1 Homo sapiens 58-62 27375190-8 2016 These results indicate that curcumin could effectively inhibit QCT induced apoptosis and inflammatory response in L02 cells, which may involve the activation of Nrf2/HO-1 and inhibition of NF-kB pathway. Curcumin 28-36 heme oxygenase 1 Homo sapiens 166-170 26938875-2 2016 Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). Carbon Monoxide 135-150 heme oxygenase 1 Homo sapiens 18-22 26938875-2 2016 Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). Carbon Monoxide 152-154 heme oxygenase 1 Homo sapiens 0-16 26938875-2 2016 Haem-oxygenase-1 (HO-1) is an enzyme responsible for the degradation of haem that can suppress inflammation, through the production of carbon monoxide (CO). Carbon Monoxide 152-154 heme oxygenase 1 Homo sapiens 18-22 27722139-0 2016 Fraxetin Induces Heme Oxygenase-1 Expression by Activation of Akt/Nrf2 or AMP-activated Protein Kinase alpha/Nrf2 Pathway in HaCaT Cells. fraxetin 0-8 heme oxygenase 1 Homo sapiens 17-33 27722139-10 2016 Pharmacological inhibition of Akt and AMPKalpha abrogated fraxetin-induced expression of HO-1 and nuclear localization of Nrf2. ampkalpha 38-47 heme oxygenase 1 Homo sapiens 89-93 27722139-10 2016 Pharmacological inhibition of Akt and AMPKalpha abrogated fraxetin-induced expression of HO-1 and nuclear localization of Nrf2. fraxetin 58-66 heme oxygenase 1 Homo sapiens 89-93 27722139-12 2016 CONCLUSIONS: Fraxetin induces HO-1 expression through activation of Akt/Nrf2 or AMPKalpha/Nrf2 pathway in HaCaT cells. fraxetin 13-21 heme oxygenase 1 Homo sapiens 30-34 27490825-6 2016 X-ray crystallographic structures of human HO1 H25R with bound heme and additional functional studies suggest that HO mutant activity inside these cells does not involve heme ligation by a proximal amino acid. Heme 63-67 heme oxygenase 1 Homo sapiens 43-46 27354356-1 2016 Epoxygenase-dependent metabolites of arachidonc acid, EETs and the heme-oxygenase (HO)-1/carbon monoxide/bilverdin system share similarities in their activity and mediators. Carbon Monoxide 89-104 heme oxygenase 1 Homo sapiens 67-88 27354356-1 2016 Epoxygenase-dependent metabolites of arachidonc acid, EETs and the heme-oxygenase (HO)-1/carbon monoxide/bilverdin system share similarities in their activity and mediators. bilverdin 105-114 heme oxygenase 1 Homo sapiens 67-88 27447561-0 2016 Targeting of heme oxygenase-1 attenuates the negative impact of Ikaros isoform 6 in adult BCR-ABL1-positive B-ALL. ikaros 64-70 heme oxygenase 1 Homo sapiens 13-29 27553905-5 2016 Western blot analysis illustrated that sulforaphane increased the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), heme oxygenase-1 (HO-1), and NAD(P)H quinone oxidoreductase (NQO1), the latter two of which are anti-oxidative enzymes. sulforaphane 39-51 heme oxygenase 1 Homo sapiens 132-148 27553905-5 2016 Western blot analysis illustrated that sulforaphane increased the expression of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), heme oxygenase-1 (HO-1), and NAD(P)H quinone oxidoreductase (NQO1), the latter two of which are anti-oxidative enzymes. sulforaphane 39-51 heme oxygenase 1 Homo sapiens 150-154 27183873-5 2016 Using the heme oxygenase-1 (HO-1) as a model of phase II enzyme gene, we found that methylation of Nrf2 by PRMT1 led to a moderate increase of its DNA-binding activity and transactivation, which subsequently protected cells against the tBHP-induced glutathione depletion and cell death. tert-Butylhydroperoxide 236-240 heme oxygenase 1 Homo sapiens 10-26 27642551-0 2016 Differential heme release from various hemoglobin redox states and the upregulation of cellular heme oxygenase-1. Heme 13-17 heme oxygenase 1 Homo sapiens 96-112 27365395-4 2016 Under endoplasmic reticulum stress conditions, induction of CSE and up-regulation of the CBS inhibitor, CO, a product of heme oxygenase-1, flip the operating preference of CSE from cystathionine to cysteine, transiently stimulating H2S production. Cystathionine 181-194 heme oxygenase 1 Homo sapiens 121-137 27365395-4 2016 Under endoplasmic reticulum stress conditions, induction of CSE and up-regulation of the CBS inhibitor, CO, a product of heme oxygenase-1, flip the operating preference of CSE from cystathionine to cysteine, transiently stimulating H2S production. Cysteine 198-206 heme oxygenase 1 Homo sapiens 121-137 27365395-4 2016 Under endoplasmic reticulum stress conditions, induction of CSE and up-regulation of the CBS inhibitor, CO, a product of heme oxygenase-1, flip the operating preference of CSE from cystathionine to cysteine, transiently stimulating H2S production. Hydrogen Sulfide 232-235 heme oxygenase 1 Homo sapiens 121-137 27183873-5 2016 Using the heme oxygenase-1 (HO-1) as a model of phase II enzyme gene, we found that methylation of Nrf2 by PRMT1 led to a moderate increase of its DNA-binding activity and transactivation, which subsequently protected cells against the tBHP-induced glutathione depletion and cell death. tert-Butylhydroperoxide 236-240 heme oxygenase 1 Homo sapiens 28-32 27183873-5 2016 Using the heme oxygenase-1 (HO-1) as a model of phase II enzyme gene, we found that methylation of Nrf2 by PRMT1 led to a moderate increase of its DNA-binding activity and transactivation, which subsequently protected cells against the tBHP-induced glutathione depletion and cell death. Glutathione 249-260 heme oxygenase 1 Homo sapiens 10-26 27183873-5 2016 Using the heme oxygenase-1 (HO-1) as a model of phase II enzyme gene, we found that methylation of Nrf2 by PRMT1 led to a moderate increase of its DNA-binding activity and transactivation, which subsequently protected cells against the tBHP-induced glutathione depletion and cell death. Glutathione 249-260 heme oxygenase 1 Homo sapiens 28-32 27190261-8 2016 These results indicated that the 1R, 1"S-isotetrandrine ameliorated tert-butyl hydroperoxide-induced oxidative damage through upregulation of heme oxygenase-1 expression by the dissociation of Nrf2 from Nrf2-Keap1 complex via extracellular signal-regulated protein kinase and c-Jun NH2-terminal kinase activation and Keap1 inactivation. tert-Butylhydroperoxide 68-92 heme oxygenase 1 Homo sapiens 142-158 26758041-3 2016 Using double immunofluorescence and sequential iron and immunohistochemistry staining, we showed that marrow iron colocalizes with HO1 and H-ferritin to CD163 + macrophages. Iron 109-113 heme oxygenase 1 Homo sapiens 131-134 27387768-9 2016 DCA and other dihydroxy-bile acids, but not PDC, induced up-regulation of both BVRalpha and heme oxygenase-1 (HO-1) in liver cells through a FXR independent and BV insensitive mechanism. Deoxycholic Acid 0-3 heme oxygenase 1 Homo sapiens 92-108 27387768-9 2016 DCA and other dihydroxy-bile acids, but not PDC, induced up-regulation of both BVRalpha and heme oxygenase-1 (HO-1) in liver cells through a FXR independent and BV insensitive mechanism. dihydroxy-bile acids 14-34 heme oxygenase 1 Homo sapiens 92-108 27494347-6 2016 Protocatechuic aldehyde exhibited a suppressive effect on intracellular reactive oxygen species generation and upregulated heme oxygenase-1 expression in Western blot analysis. Aldehydes 15-23 heme oxygenase 1 Homo sapiens 123-139 27190261-0 2016 Isotetrandrine ameliorates tert-butyl hydroperoxide-induced oxidative stress through upregulation of heme oxygenase-1 expression. tetrandrine 0-14 heme oxygenase 1 Homo sapiens 101-117 27190261-0 2016 Isotetrandrine ameliorates tert-butyl hydroperoxide-induced oxidative stress through upregulation of heme oxygenase-1 expression. tert-Butylhydroperoxide 27-51 heme oxygenase 1 Homo sapiens 101-117 27190261-4 2016 Additionally, our study confirmed that 1R, 1"S-isotetrandrine significantly increased the antioxidant enzyme heme oxygenase-1 expression and nuclear translocation of factor-erythroid 2 p45-related factor 2 (Nrf2). 1r, 1"s-isotetrandrine 39-61 heme oxygenase 1 Homo sapiens 109-125 27190261-8 2016 These results indicated that the 1R, 1"S-isotetrandrine ameliorated tert-butyl hydroperoxide-induced oxidative damage through upregulation of heme oxygenase-1 expression by the dissociation of Nrf2 from Nrf2-Keap1 complex via extracellular signal-regulated protein kinase and c-Jun NH2-terminal kinase activation and Keap1 inactivation. 1r, 1"s-isotetrandrine 33-55 heme oxygenase 1 Homo sapiens 142-158 26758041-1 2016 Iron overload and transfusion dependance portend poor risk in myelodysplastic syndromes (MDS); bone marrow macrophages store iron and limit oxidative damage through heme oxygenase-1 (HO1). Iron 0-4 heme oxygenase 1 Homo sapiens 183-186 26758041-1 2016 Iron overload and transfusion dependance portend poor risk in myelodysplastic syndromes (MDS); bone marrow macrophages store iron and limit oxidative damage through heme oxygenase-1 (HO1). Iron 125-129 heme oxygenase 1 Homo sapiens 183-186 26966892-6 2016 Zinc protoporphyrin IX, a specific competitive inhibitor of HO-1, efficiently inhibited RAR whereas hemin, an inducer of HO-1, could mimic priming dose of X-rays to induce RAR. zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 60-64 26795735-6 2016 Inhibition of TrxR using lower auranofin concentrations induced HO-1 protein expression in myeloma cells. Auranofin 31-40 heme oxygenase 1 Homo sapiens 64-68 27444578-6 2016 Molecular data revealed that Zn-doped TiO2 NPs induced the down-regulation of super oxide dismutase gene while the up-regulation of heme oxygenase-1 gene in MCF-7 cells. Zinc 29-31 heme oxygenase 1 Homo sapiens 132-148 26795735-8 2016 TrxR was shown to regulate HO-1 via the Nrf2 signaling pathway in a ROS-dependent manner. ros 68-71 heme oxygenase 1 Homo sapiens 27-31 26795735-9 2016 Increased HO-1 mRNA levels were observed in bortezomib-resistant myeloma cells compared to parent cells and HO-1 inhibition restored the sensitivity to bortezomib in bortezomib-resistant myeloma cells. Bortezomib 44-54 heme oxygenase 1 Homo sapiens 10-14 26795735-9 2016 Increased HO-1 mRNA levels were observed in bortezomib-resistant myeloma cells compared to parent cells and HO-1 inhibition restored the sensitivity to bortezomib in bortezomib-resistant myeloma cells. Bortezomib 152-162 heme oxygenase 1 Homo sapiens 108-112 26795735-9 2016 Increased HO-1 mRNA levels were observed in bortezomib-resistant myeloma cells compared to parent cells and HO-1 inhibition restored the sensitivity to bortezomib in bortezomib-resistant myeloma cells. Bortezomib 152-162 heme oxygenase 1 Homo sapiens 108-112 26795735-10 2016 These findings indicate that concurrent inhibition of HO-1 with either a TrxR inhibitor or with bortezomib would improve therapeutic outcomes in MM patients. Bortezomib 96-106 heme oxygenase 1 Homo sapiens 54-58 27444578-6 2016 Molecular data revealed that Zn-doped TiO2 NPs induced the down-regulation of super oxide dismutase gene while the up-regulation of heme oxygenase-1 gene in MCF-7 cells. titanium dioxide 38-42 heme oxygenase 1 Homo sapiens 132-148 26991755-6 2016 Up-regulation of HMOX1, a key regulator of cellular response to ROS, was independent of TP53 mutational status. Reactive Oxygen Species 64-67 heme oxygenase 1 Homo sapiens 17-22 27477937-11 2016 Upregulation of HO-1 by cobalt-protoporphyrin treatment increased the osteogenic differentiation of pMSCs under hypoxia, while inhibition of HO-1 by Zn-protoporphyrin reduced the osteogenic differentiation of hypoxic pMSCs. cobaltiprotoporphyrin 24-45 heme oxygenase 1 Homo sapiens 16-20 27477937-11 2016 Upregulation of HO-1 by cobalt-protoporphyrin treatment increased the osteogenic differentiation of pMSCs under hypoxia, while inhibition of HO-1 by Zn-protoporphyrin reduced the osteogenic differentiation of hypoxic pMSCs. zn-protoporphyrin 149-166 heme oxygenase 1 Homo sapiens 141-145 27323826-0 2016 Simvastatin induces heme oxygenase-1 via NF-E2-related factor 2 (Nrf2) activation through ERK and PI3K/Akt pathway in colon cancer. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 20-36 27323826-6 2016 We evaluated which signal pathway such as ERK or PI3K pathway affect Nrf2 activation and whether simvastatin induces antioxidant enzymes (heme oxygenase-1 (HO-1), NAD(P)H: quinine oxidoreductase 1 (NQO1), gamma-glutamate-cysteine ligase catalytic subunit (GCLC)). Simvastatin 97-108 heme oxygenase 1 Homo sapiens 138-154 27323826-8 2016 We also demonstrated that simvastatin-induced anti-oxidant enzymes (HO-1, NQO1, and GCLC) in HT-29 and HCT 116 cells. Simvastatin 26-37 heme oxygenase 1 Homo sapiens 68-72 27323826-9 2016 PI3K/Akt inhibitor (LY294002) and ERK inhibitor (PD98059) suppressed simvastatin-induced Nrf2 and HO-1 expression in both HT-29 and HCT 116 cells. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 20-28 heme oxygenase 1 Homo sapiens 98-102 27323826-9 2016 PI3K/Akt inhibitor (LY294002) and ERK inhibitor (PD98059) suppressed simvastatin-induced Nrf2 and HO-1 expression in both HT-29 and HCT 116 cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 49-56 heme oxygenase 1 Homo sapiens 98-102 27323826-9 2016 PI3K/Akt inhibitor (LY294002) and ERK inhibitor (PD98059) suppressed simvastatin-induced Nrf2 and HO-1 expression in both HT-29 and HCT 116 cells. Simvastatin 69-80 heme oxygenase 1 Homo sapiens 98-102 27381978-7 2016 In the human endothelial Ea.hy926 cell line, we demonstrated that intracellular bilirubin (3-5 pmol mg(-1) protein) could be modulated by either extracellular bilirubin uptake, or by up-regulation of heme oxygenase-1, a cellular enzyme related to endogenous bilirubin synthesis. Bilirubin 80-89 heme oxygenase 1 Homo sapiens 200-216 26991755-10 2016 In conclusion, ROS induction is important for antileukaemic activities of APR-246 and inhibiting the protective response of the Nrf-2/HMOX1 axis using PI3K inhibitors, enhances the antileukaemic effects. Reactive Oxygen Species 15-18 heme oxygenase 1 Homo sapiens 134-139 27381601-0 2016 Simvastatin Inhibits Epithelial-to-Mesenchymal Transition Through Induction of HO-1 in Cultured Renal Proximal Tubule Cells. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 79-83 27317992-5 2016 In addition, dioscin markedly up-regulated the expression levels of SIRT1, HO-1, NQO1, GST and GCLM through increasing nuclear translocation of Nrf2 against oxidative stress. dioscin 13-20 heme oxygenase 1 Homo sapiens 75-79 27107768-0 2016 Rupestonic acid derivative YZH-106 suppresses influenza virus replication by activation of heme oxygenase-1-mediated interferon response. rupestonic acid 0-15 heme oxygenase 1 Homo sapiens 91-107 27107768-0 2016 Rupestonic acid derivative YZH-106 suppresses influenza virus replication by activation of heme oxygenase-1-mediated interferon response. YZH-106 27-34 heme oxygenase 1 Homo sapiens 91-107 27107768-6 2016 Mechanistically, YZH-106 induced p38 MAPK and ERK1/2 phosphorylation, which led to the activation of erythroid 2-related factor 2 (Nrf2) that up-regulated heme oxygenase-1 (HO-1) expression in addition to other genes. YZH-106 17-24 heme oxygenase 1 Homo sapiens 155-171 27107768-6 2016 Mechanistically, YZH-106 induced p38 MAPK and ERK1/2 phosphorylation, which led to the activation of erythroid 2-related factor 2 (Nrf2) that up-regulated heme oxygenase-1 (HO-1) expression in addition to other genes. YZH-106 17-24 heme oxygenase 1 Homo sapiens 173-177 27107768-8 2016 These results suggest that YZH-106 inhibits IAV by up-regulating HO-1-mediated IFN response. YZH-106 27-34 heme oxygenase 1 Homo sapiens 65-69 26588813-8 2016 These results were validated by quantitative reverse transcriptase-polymerase chain reaction (RT-PCR) and Western blotting, confirming a concentration-dependent increase in HMOX1 and a decrease in THBS1 expression in ASC following iAs exposure. 4-Iodoacetamidosalicylic acid 231-234 heme oxygenase 1 Homo sapiens 173-178 27381601-1 2016 BACKGROUND/AIM: Studies have shown that simvastatin (SIM) inhibits epithelial-mesenchymal transition (EMT), a key step in fibrosis, and activates the anti-fibrotic heme oxygenase-1 (HO-1) gene in renal proximal tubule cells independent of its lipid-lowering. Simvastatin 40-51 heme oxygenase 1 Homo sapiens 164-180 27381601-1 2016 BACKGROUND/AIM: Studies have shown that simvastatin (SIM) inhibits epithelial-mesenchymal transition (EMT), a key step in fibrosis, and activates the anti-fibrotic heme oxygenase-1 (HO-1) gene in renal proximal tubule cells independent of its lipid-lowering. Simvastatin 40-51 heme oxygenase 1 Homo sapiens 182-186 27381601-2 2016 We tested the hypothesis that SIM inhibits EMT via HO-1-dependent suppression of reactive oxygen species (ROS) release. Reactive Oxygen Species 81-104 heme oxygenase 1 Homo sapiens 51-55 27381601-2 2016 We tested the hypothesis that SIM inhibits EMT via HO-1-dependent suppression of reactive oxygen species (ROS) release. Reactive Oxygen Species 106-109 heme oxygenase 1 Homo sapiens 51-55 27381601-7 2016 CONCLUSION: SIM, via HO-1, suppresses TGFbeta1-dependent ROS production and, hence, EMT. Reactive Oxygen Species 57-60 heme oxygenase 1 Homo sapiens 21-25 27151894-11 2016 Expression of some key oxidative stress-responsive genes, such as heme oxygenase-1 and metallothionein-1, was also stimulated by iAs exposure. 4-Iodoacetamidosalicylic acid 129-132 heme oxygenase 1 Homo sapiens 66-82 26969214-0 2016 Aspirin induces Nrf2-mediated transcriptional activation of haem oxygenase-1 in protection of human melanocytes from H2 O2 -induced oxidative stress. Aspirin 0-7 heme oxygenase 1 Homo sapiens 60-76 26969214-0 2016 Aspirin induces Nrf2-mediated transcriptional activation of haem oxygenase-1 in protection of human melanocytes from H2 O2 -induced oxidative stress. Hydrogen Peroxide 117-122 heme oxygenase 1 Homo sapiens 60-76 26969214-9 2016 Furthermore, we found ASA dramatically induced NRF2 nuclear translocation, enhanced ARE-luciferase activity, increased both p- NRF2 and total NRF2 levels, and induced the expression of haem oxygenase-1 (HO-1) in human melanocytes. Aspirin 22-25 heme oxygenase 1 Homo sapiens 185-201 26969214-9 2016 Furthermore, we found ASA dramatically induced NRF2 nuclear translocation, enhanced ARE-luciferase activity, increased both p- NRF2 and total NRF2 levels, and induced the expression of haem oxygenase-1 (HO-1) in human melanocytes. Aspirin 22-25 heme oxygenase 1 Homo sapiens 203-207 26969214-10 2016 In addition, knockdown of Nrf2 expression or pharmacological inhibition of HO-1 abrogated the protective action of ASA on melanocytes against H2 O2 -induced cytotoxicity and apoptosis. Aspirin 115-118 heme oxygenase 1 Homo sapiens 75-79 26969214-10 2016 In addition, knockdown of Nrf2 expression or pharmacological inhibition of HO-1 abrogated the protective action of ASA on melanocytes against H2 O2 -induced cytotoxicity and apoptosis. Hydrogen Peroxide 142-147 heme oxygenase 1 Homo sapiens 75-79 26969214-11 2016 These results suggest that ASA protects human melanocytes against H2 O2 -induced oxidative stress via Nrf2-driven transcriptional activation of HO-1. Aspirin 27-30 heme oxygenase 1 Homo sapiens 144-148 26969214-11 2016 These results suggest that ASA protects human melanocytes against H2 O2 -induced oxidative stress via Nrf2-driven transcriptional activation of HO-1. Hydrogen Peroxide 66-71 heme oxygenase 1 Homo sapiens 144-148 27176599-0 2016 In vivo response of heme-oxygenase-1 to metal ions released from metal-on-metal hip prostheses. Metals 40-45 heme oxygenase 1 Homo sapiens 20-36 27211510-9 2016 Combination of the NF-kappaB inhibitor Bay11-7082 and the lentivirus vector Lenti-siHO-1 significantly decreased HO-1 protein expression and increased apoptosis in OCI-ly10 cells. 3-(4-methylphenylsulfonyl)-2-propenenitrile 39-49 heme oxygenase 1 Homo sapiens 84-88 27087120-8 2016 SHSY5Y cells treated with quercetin also increased the expression of Nrf2 (via a decrease in the levels of Keap1), heme oxygenase-1 (HO-1), and nitric oxide synthase 1 (NOS1), which provide further protection from oxidative stress. Quercetin 26-35 heme oxygenase 1 Homo sapiens 115-131 27087120-8 2016 SHSY5Y cells treated with quercetin also increased the expression of Nrf2 (via a decrease in the levels of Keap1), heme oxygenase-1 (HO-1), and nitric oxide synthase 1 (NOS1), which provide further protection from oxidative stress. Quercetin 26-35 heme oxygenase 1 Homo sapiens 133-137 27087120-10 2016 We hypothesize that SUMOylated HIF-1alpha plays a fundamental role in the protection afforded and may underlie some of quercetin"s ability to protect cells from oxygen/glucose deprivation-induced cell death, via an up-regulation of HO-1 and NOS1, which ultimately leads to the induction of pro-life NOS1/protein kinase G signaling. Quercetin 119-128 heme oxygenase 1 Homo sapiens 232-236 27206323-8 2016 NO2 elicited the greatest HMOX1 response, whereas O3 more greatly induced IL-6, IL-8 and PTGS2 expression. Nitrogen Dioxide 0-3 heme oxygenase 1 Homo sapiens 26-31 27176599-0 2016 In vivo response of heme-oxygenase-1 to metal ions released from metal-on-metal hip prostheses. Metals 65-70 heme oxygenase 1 Homo sapiens 20-36 27176599-3 2016 The aim of the present study was to verify whether, in patients with a MoM hip prosthesis, mRNA and protein expression of HMOX-1 was modulated by the presence of metal ions and whether patients without prostheses exhibit a different expression pattern of this enzyme. Metals 162-167 heme oxygenase 1 Homo sapiens 122-128 28933402-5 2016 Although other chemopreventive mechanisms have been identified, the primary mechanism believed to be responsible for the observed chemoprevention from GSHPs is the induction of antioxidant enzymes, such as NAD(P)H quinone reductase (NQO1), heme oxygenase 1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glutathione S transferases (GSTs), through the Keap1-Nrf2-ARE signaling pathway. gshps 151-156 heme oxygenase 1 Homo sapiens 240-256 27176653-9 2016 A series of increased oxidative stress toxicological responses, including cellular ROS production, heme oxygenase-1 expression, cellular GSH depletion, and mitochondrial dysfunctions, were triggered by faceted TiO2 nanocrystals with progressively increased {001} percentages, demonstrating the toxicological roles of {001} facets. titanium dioxide 210-214 heme oxygenase 1 Homo sapiens 99-115 28933402-5 2016 Although other chemopreventive mechanisms have been identified, the primary mechanism believed to be responsible for the observed chemoprevention from GSHPs is the induction of antioxidant enzymes, such as NAD(P)H quinone reductase (NQO1), heme oxygenase 1 (HO-1), glutamate-cysteine ligase catalytic subunit (GCLC), and glutathione S transferases (GSTs), through the Keap1-Nrf2-ARE signaling pathway. gshps 151-156 heme oxygenase 1 Homo sapiens 258-262 27122182-1 2016 Increasing evidence has demonstrated that the activation of heme oxygenase (HO)-1 reduces autophagy stimulated by oxidative stress injury, in which the supraphysiological production of reactive oxygen species (ROS) is detected. Reactive Oxygen Species 185-208 heme oxygenase 1 Homo sapiens 60-81 27142241-5 2016 Many nuclear processes are influenced by a spatial switch involving the proteins {KPNA2, KPNB1, PCNA, PTMA, SET} and heme/iron proteins HMOX1 and FTH1. Iron 122-126 heme oxygenase 1 Homo sapiens 136-141 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. Bromine 64-67 heme oxygenase 1 Homo sapiens 106-122 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. Bromine 64-67 heme oxygenase 1 Homo sapiens 124-128 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. Heme 84-88 heme oxygenase 1 Homo sapiens 106-122 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. Heme 84-88 heme oxygenase 1 Homo sapiens 124-128 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. Heme 106-110 heme oxygenase 1 Homo sapiens 124-128 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. bilverdin 162-171 heme oxygenase 1 Homo sapiens 106-122 27244263-4 2016 Our study was based on previous findings that demonstrated that Br2 upregulates the heme-degrading enzyme heme oxygenase-1 (HO-1), which converts toxic heme into bilverdin. bilverdin 162-171 heme oxygenase 1 Homo sapiens 124-128 27244263-7 2016 However, therapeutic reduction of heme levels, by either scavenging with hemopexin or degradation by HO-1, improved lung function and survival. Heme 34-38 heme oxygenase 1 Homo sapiens 101-105 27044026-2 2016 The inducible enzyme heme oxygenase-1 (HO-1) protects cells against inflammation and can be induced by electrophilic compounds like the chalcones (1,3-diphenylprop-2-enones) from the class of alpha,beta-unsaturated carbonyl compounds. Chalcones 136-145 heme oxygenase 1 Homo sapiens 39-43 27044026-2 2016 The inducible enzyme heme oxygenase-1 (HO-1) protects cells against inflammation and can be induced by electrophilic compounds like the chalcones (1,3-diphenylprop-2-enones) from the class of alpha,beta-unsaturated carbonyl compounds. 1,3-diphenylprop-2-enones 147-172 heme oxygenase 1 Homo sapiens 39-43 27044026-2 2016 The inducible enzyme heme oxygenase-1 (HO-1) protects cells against inflammation and can be induced by electrophilic compounds like the chalcones (1,3-diphenylprop-2-enones) from the class of alpha,beta-unsaturated carbonyl compounds. alpha,beta-unsaturated carbonyl compounds 192-233 heme oxygenase 1 Homo sapiens 39-43 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Heme 170-174 heme oxygenase 1 Homo sapiens 188-192 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Carbon Monoxide 283-298 heme oxygenase 1 Homo sapiens 170-186 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Carbon Monoxide 283-298 heme oxygenase 1 Homo sapiens 188-192 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Biliverdine 300-310 heme oxygenase 1 Homo sapiens 170-186 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Biliverdine 300-310 heme oxygenase 1 Homo sapiens 188-192 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Iron 316-320 heme oxygenase 1 Homo sapiens 170-186 27283533-4 2016 Although the antioxidant and heat-shock proteins are included in this category, one enzyme that has received a great deal of attention as a master protective sentinel is heme oxygenase-1 (HO-1), the rate-limiting step in the catabolism of heme into the bioactive signaling molecules carbon monoxide, biliverdin, and iron. Iron 316-320 heme oxygenase 1 Homo sapiens 188-192 26899309-7 2016 We further demonstrated the antioxidant effect of tangeretin by showing that tangeretin inhibited reactive oxygen species production and p47(phox) phosphorylation, while enhancing the expression of heme oxygenase-1 and the DNA binding activity of nuclear factor-erythroid 2-related factor 2 to the antioxidant response element in LPS-stimulated microglia. tangeretin 50-60 heme oxygenase 1 Homo sapiens 198-214 26899309-7 2016 We further demonstrated the antioxidant effect of tangeretin by showing that tangeretin inhibited reactive oxygen species production and p47(phox) phosphorylation, while enhancing the expression of heme oxygenase-1 and the DNA binding activity of nuclear factor-erythroid 2-related factor 2 to the antioxidant response element in LPS-stimulated microglia. tangeretin 77-87 heme oxygenase 1 Homo sapiens 198-214 27122182-1 2016 Increasing evidence has demonstrated that the activation of heme oxygenase (HO)-1 reduces autophagy stimulated by oxidative stress injury, in which the supraphysiological production of reactive oxygen species (ROS) is detected. Reactive Oxygen Species 210-213 heme oxygenase 1 Homo sapiens 60-81 26930712-0 2016 Heme oxygenase-1 nuclear translocation regulates bortezomibinduced cytotoxicity and mediates genomic instability in myeloma cells. bortezomibinduced 49-66 heme oxygenase 1 Homo sapiens 0-16 27198537-2 2016 After ICH, overproduction of iron associated with induction of heme oxygenase-1 (HO-1) in brain was observed. Iron 29-33 heme oxygenase 1 Homo sapiens 63-79 27198537-2 2016 After ICH, overproduction of iron associated with induction of heme oxygenase-1 (HO-1) in brain was observed. Iron 29-33 heme oxygenase 1 Homo sapiens 81-85 27198537-5 2016 The present results showed a higher level of HO-1 expression associated with more severe injury in males compared with females after FC-infusion. ferrous citrate 133-135 heme oxygenase 1 Homo sapiens 45-49 27198537-11 2016 These findings open up the prospect for male-specific neuroprotection targeting HO-1 suppression for patients suffering from striatal iron overload. Iron 134-138 heme oxygenase 1 Homo sapiens 80-84 27035791-11 2016 We also demonstrated that Rev-AuNPs induced heme oxygenase-1 (HO-1) expression and that the inhibition of MMP-9 and COX-2 expression and MMP-9 enzymatic activity of Rev-AuNPs were abrogated by siRNA knockdown of HO-1 expression. rev-aunps 26-35 heme oxygenase 1 Homo sapiens 44-60 27035791-11 2016 We also demonstrated that Rev-AuNPs induced heme oxygenase-1 (HO-1) expression and that the inhibition of MMP-9 and COX-2 expression and MMP-9 enzymatic activity of Rev-AuNPs were abrogated by siRNA knockdown of HO-1 expression. rev-aunps 26-35 heme oxygenase 1 Homo sapiens 62-66 27035791-11 2016 We also demonstrated that Rev-AuNPs induced heme oxygenase-1 (HO-1) expression and that the inhibition of MMP-9 and COX-2 expression and MMP-9 enzymatic activity of Rev-AuNPs were abrogated by siRNA knockdown of HO-1 expression. rev-aunps 165-174 heme oxygenase 1 Homo sapiens 44-60 27035791-11 2016 We also demonstrated that Rev-AuNPs induced heme oxygenase-1 (HO-1) expression and that the inhibition of MMP-9 and COX-2 expression and MMP-9 enzymatic activity of Rev-AuNPs were abrogated by siRNA knockdown of HO-1 expression. rev-aunps 165-174 heme oxygenase 1 Homo sapiens 62-66 27035791-11 2016 We also demonstrated that Rev-AuNPs induced heme oxygenase-1 (HO-1) expression and that the inhibition of MMP-9 and COX-2 expression and MMP-9 enzymatic activity of Rev-AuNPs were abrogated by siRNA knockdown of HO-1 expression. rev-aunps 165-174 heme oxygenase 1 Homo sapiens 212-216 27035791-12 2016 Our findings revealed that the anti-invasive effects of Rev-AuNPs in response to TPA-stimulation were mediated by the suppression of MMP-9, COX-2, NF-kappaB, AP-1, PI3K/Akt and ERK and/or the activation of HO-1 signaling cascades. rev-aunps 56-65 heme oxygenase 1 Homo sapiens 206-210 27035791-12 2016 Our findings revealed that the anti-invasive effects of Rev-AuNPs in response to TPA-stimulation were mediated by the suppression of MMP-9, COX-2, NF-kappaB, AP-1, PI3K/Akt and ERK and/or the activation of HO-1 signaling cascades. Tetradecanoylphorbol Acetate 81-84 heme oxygenase 1 Homo sapiens 206-210 26930712-3 2016 The aim of the present research was to evaluate the impact of HO-1 in MM following bortezomib (BTZ) treatment and how HO-1 is implicated in the mechanisms of chemoresistance. Bortezomib 83-93 heme oxygenase 1 Homo sapiens 62-66 26930712-3 2016 The aim of the present research was to evaluate the impact of HO-1 in MM following bortezomib (BTZ) treatment and how HO-1 is implicated in the mechanisms of chemoresistance. Bortezomib 95-98 heme oxygenase 1 Homo sapiens 62-66 26930712-10 2016 In conclusion, our data suggest that BTZ sensitivity depends on HO-1 nuclear compartmentalization and not on its enzymatic activity and this finding may represent an important tool to overcome BTZ chemoresistance in MM patients. Bortezomib 37-40 heme oxygenase 1 Homo sapiens 64-68 27059143-0 2016 Methionine sulfoxide reductase B3 deficiency stimulates heme oxygenase-1 expression via ROS-dependent and Nrf2 activation pathways. Reactive Oxygen Species 88-91 heme oxygenase 1 Homo sapiens 56-72 27059143-10 2016 Taken together, these data suggest that MsrB3 attenuates HO-1 induction by inhibiting ROS production, ER stress, and Nrf2 activation. Reactive Oxygen Species 86-89 heme oxygenase 1 Homo sapiens 57-61 27059143-7 2016 Treatment with N-acetylcysteine as an ROS scavenger reduced augmented HO-1 levels in MsrB3-depleted cells. Acetylcysteine 15-31 heme oxygenase 1 Homo sapiens 70-74 27059143-7 2016 Treatment with N-acetylcysteine as an ROS scavenger reduced augmented HO-1 levels in MsrB3-depleted cells. Reactive Oxygen Species 38-41 heme oxygenase 1 Homo sapiens 70-74 27094403-9 2016 The pretreatments with monohydroxy-DMC and monohydroxy-BDMC reduced c-jun and c-fos mRNA expression and p53 tumor suppressor protein expression and increased HO-1 protein expression and glutathione peroxidase (GPx) activity, respectively, compared to cells with direct hydrogen peroxide treatments. monohydroxy-dmc 23-38 heme oxygenase 1 Homo sapiens 158-162 27005776-7 2016 MT protein was further induced by as little as 0.5muM cadmium, reaching a 6-fold induction at 20muM, whereas for HO-1, a 5muM cadmium concentration was required for initial induction and at 20muM cadmium reached a 15-fold induction. Cadmium 126-133 heme oxygenase 1 Homo sapiens 113-117 27005776-7 2016 MT protein was further induced by as little as 0.5muM cadmium, reaching a 6-fold induction at 20muM, whereas for HO-1, a 5muM cadmium concentration was required for initial induction and at 20muM cadmium reached a 15-fold induction. Cadmium 126-133 heme oxygenase 1 Homo sapiens 113-117 27005776-10 2016 In conclusion although cultured PTC, do express CYP proteins, (CYP2E1, CYP3A4, and CYP4F2), Cd-induced cell stress as indicted by induction of HO-1 and MT does not alter expression of these CYP proteins at 48h. Cadmium 92-94 heme oxygenase 1 Homo sapiens 143-154 27094403-9 2016 The pretreatments with monohydroxy-DMC and monohydroxy-BDMC reduced c-jun and c-fos mRNA expression and p53 tumor suppressor protein expression and increased HO-1 protein expression and glutathione peroxidase (GPx) activity, respectively, compared to cells with direct hydrogen peroxide treatments. monohydroxy-bdmc 43-59 heme oxygenase 1 Homo sapiens 158-162 26795536-9 2016 In addition, Trp increased the expression of the Nrf2-dependent antioxidant genes NQO1, HO-1 and GCS in hepatocytes both in vitro and in vivo. Tryptophan 13-16 heme oxygenase 1 Homo sapiens 88-92 27164116-6 2016 Furthermore, DMSP was able to prevent the cytotoxic effects exerted by TDA, including the breakdown of the mitochondrial membrane potential, upregulation of heat shock protein Hsp32 and activation of the extracellular signal-regulated kinases 1/2 (ERK1/2). dimethylpropiothetin 13-17 heme oxygenase 1 Homo sapiens 176-181 26889770-3 2016 Our results showed that pretreatment with l-carnitine augmented Nrf2 nuclear translocation, DNA binding activity and heme oxygenase-1 (HO-1) expression in H2O2-treated HL7702 cells, although l-carnitine treatment alone had no effect on them. Carnitine 42-53 heme oxygenase 1 Homo sapiens 117-133 26889770-3 2016 Our results showed that pretreatment with l-carnitine augmented Nrf2 nuclear translocation, DNA binding activity and heme oxygenase-1 (HO-1) expression in H2O2-treated HL7702 cells, although l-carnitine treatment alone had no effect on them. Carnitine 42-53 heme oxygenase 1 Homo sapiens 135-139 26889770-4 2016 Analysis using Nrf2 siRNA demonstrated that Nrf2 activation was involved in l-carnitine-induced HO-1 expression. Carnitine 76-87 heme oxygenase 1 Homo sapiens 96-100 26889770-8 2016 Moreover, our finding demonstrated that the induction of Nrf2 translocation and HO-1 expression by l-carnitine directly correlated with the Akt pathway because Akt inhibitor showed inhibitory effects on the Nrf2 translocation and HO-1 expression. Carnitine 99-110 heme oxygenase 1 Homo sapiens 80-84 26889770-8 2016 Moreover, our finding demonstrated that the induction of Nrf2 translocation and HO-1 expression by l-carnitine directly correlated with the Akt pathway because Akt inhibitor showed inhibitory effects on the Nrf2 translocation and HO-1 expression. Carnitine 99-110 heme oxygenase 1 Homo sapiens 230-234 26889770-3 2016 Our results showed that pretreatment with l-carnitine augmented Nrf2 nuclear translocation, DNA binding activity and heme oxygenase-1 (HO-1) expression in H2O2-treated HL7702 cells, although l-carnitine treatment alone had no effect on them. Hydrogen Peroxide 155-159 heme oxygenase 1 Homo sapiens 117-133 26889770-3 2016 Our results showed that pretreatment with l-carnitine augmented Nrf2 nuclear translocation, DNA binding activity and heme oxygenase-1 (HO-1) expression in H2O2-treated HL7702 cells, although l-carnitine treatment alone had no effect on them. Hydrogen Peroxide 155-159 heme oxygenase 1 Homo sapiens 135-139 26878775-4 2016 ECG attenuated lipopolysaccharide (LPS)-induced inflammatory mediator expression and intracellular reactive oxygen species (ROS) generation through the induction of Nrf2/antioxidant response element (ARE)-driven glutathione (GSH) and hemeoxygenase-1 (HO-1) levels, interference with NF-kappaB and Nfr2/ARE transcriptional activities, and suppression of the MAPKs (JNK1/2 and p38) and PI3K/Akt signaling pathways. epicatechin gallate 0-3 heme oxygenase 1 Homo sapiens 234-255 26992777-0 2016 Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Hydrogen Peroxide 80-97 heme oxygenase 1 Homo sapiens 0-16 26992777-0 2016 Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Hydrogen Peroxide 80-97 heme oxygenase 1 Homo sapiens 18-22 26992777-0 2016 Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Hydrogen Peroxide 99-103 heme oxygenase 1 Homo sapiens 0-16 26992777-0 2016 Heme oxygenase-1 (HO-1) protects human lens epithelial cells (SRA01/04) against hydrogen peroxide (H2O2)-induced oxidative stress and apoptosis. Hydrogen Peroxide 99-103 heme oxygenase 1 Homo sapiens 18-22 26992777-1 2016 OBJECTIVES: This study aimed to investigate the protective role of heme oxygenase-1 (HO-1) in H2O2-induced oxidative stress and apoptosis in human lens epithelial cells (hLEC; SRA01/04). Hydrogen Peroxide 94-98 heme oxygenase 1 Homo sapiens 67-83 26992777-1 2016 OBJECTIVES: This study aimed to investigate the protective role of heme oxygenase-1 (HO-1) in H2O2-induced oxidative stress and apoptosis in human lens epithelial cells (hLEC; SRA01/04). Hydrogen Peroxide 94-98 heme oxygenase 1 Homo sapiens 85-89 26992777-12 2016 RESULTS: HO-1 significantly restored the cell viability under H2O2 injury via reducing the generation of ROS and increasing the levels of SOD and GSH activity. Hydrogen Peroxide 62-66 heme oxygenase 1 Homo sapiens 9-13 26992777-12 2016 RESULTS: HO-1 significantly restored the cell viability under H2O2 injury via reducing the generation of ROS and increasing the levels of SOD and GSH activity. Reactive Oxygen Species 105-108 heme oxygenase 1 Homo sapiens 9-13 26992777-12 2016 RESULTS: HO-1 significantly restored the cell viability under H2O2 injury via reducing the generation of ROS and increasing the levels of SOD and GSH activity. Glutathione 146-149 heme oxygenase 1 Homo sapiens 9-13 26968795-0 2016 Heme oxygenase-1-derived bilirubin counteracts HIV protease inhibitor-mediated endothelial cell dysfunction. Bilirubin 25-34 heme oxygenase 1 Homo sapiens 0-16 26992777-13 2016 Moreover, HO-1 also inhibited H2O2-induced caspase-8 and caspase-3 proteins, thus significantly reducing the apoptosis of SRA01/04. Hydrogen Peroxide 30-34 heme oxygenase 1 Homo sapiens 10-14 26878775-4 2016 ECG attenuated lipopolysaccharide (LPS)-induced inflammatory mediator expression and intracellular reactive oxygen species (ROS) generation through the induction of Nrf2/antioxidant response element (ARE)-driven glutathione (GSH) and hemeoxygenase-1 (HO-1) levels, interference with NF-kappaB and Nfr2/ARE transcriptional activities, and suppression of the MAPKs (JNK1/2 and p38) and PI3K/Akt signaling pathways. Glutathione 212-223 heme oxygenase 1 Homo sapiens 234-255 26992777-15 2016 CONCLUSIONS: These findings suggested that HO-1 protects human lens epithelial cells from H2O2-induced oxidant stress by upregulating antioxidant enzyme activity, reducing ROS generation, and thus inhibiting caspase family-dependent apoptosis. Hydrogen Peroxide 90-94 heme oxygenase 1 Homo sapiens 43-47 26992777-15 2016 CONCLUSIONS: These findings suggested that HO-1 protects human lens epithelial cells from H2O2-induced oxidant stress by upregulating antioxidant enzyme activity, reducing ROS generation, and thus inhibiting caspase family-dependent apoptosis. Reactive Oxygen Species 172-175 heme oxygenase 1 Homo sapiens 43-47 26968795-4 2016 We found that three distinct PIs, including ritonavir, atazanavir, and lopinavir, stimulated the expression of HO-1 protein and mRNA. Ritonavir 44-53 heme oxygenase 1 Homo sapiens 111-115 26968795-4 2016 We found that three distinct PIs, including ritonavir, atazanavir, and lopinavir, stimulated the expression of HO-1 protein and mRNA. Atazanavir Sulfate 55-65 heme oxygenase 1 Homo sapiens 111-115 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 32-56 heme oxygenase 1 Homo sapiens 143-147 26968795-4 2016 We found that three distinct PIs, including ritonavir, atazanavir, and lopinavir, stimulated the expression of HO-1 protein and mRNA. Lopinavir 71-80 heme oxygenase 1 Homo sapiens 111-115 26968795-5 2016 The induction of HO-1 was associated with an increase in NF-E2-related factor-2 (Nrf2) activity and reactive oxygen species (ROS). Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 17-21 26968795-5 2016 The induction of HO-1 was associated with an increase in NF-E2-related factor-2 (Nrf2) activity and reactive oxygen species (ROS). Reactive Oxygen Species 125-128 heme oxygenase 1 Homo sapiens 17-21 26968795-7 2016 In addition, the PI-mediated induction of HO-1 was abolished by N-acetyl-l-cysteine and rotenone. Acetylcysteine 64-83 heme oxygenase 1 Homo sapiens 42-46 26968795-7 2016 In addition, the PI-mediated induction of HO-1 was abolished by N-acetyl-l-cysteine and rotenone. Rotenone 88-96 heme oxygenase 1 Homo sapiens 42-46 26968795-9 2016 Inhibition of HO-1 activity or expression potentiated the anti-proliferative and inflammatory actions of PIs which was reversed by bilirubin but not carbon monoxide. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 14-18 26968795-12 2016 Thus, induction of HO-1 via the ROS-Nrf2 pathway in human ECs counteracts the anti-proliferative and inflammatory actions of PIs by generating bilirubin. Reactive Oxygen Species 32-35 heme oxygenase 1 Homo sapiens 19-23 26968795-12 2016 Thus, induction of HO-1 via the ROS-Nrf2 pathway in human ECs counteracts the anti-proliferative and inflammatory actions of PIs by generating bilirubin. Bilirubin 143-152 heme oxygenase 1 Homo sapiens 19-23 27124156-5 2016 In vitro study showed that OSCC cell lines, especially HO1-N-1 cells stimulated NOTCH3 expression in normal human dermal fibroblasts (NHDFs) through direct cell-to-cell contact. nhdfs 134-139 heme oxygenase 1 Homo sapiens 55-58 26993595-4 2016 We show that the effects of CO on tumor stroma and reprogramming of macrophages towards the anti-tumoral phenotype is mediated by reactive oxygen species (ROS)-dependent activation of MAPK/Erk1/2-c-myc pathway as well as Notch 1-dependent negative feedback on the metabolic enzyme heme oxygenase-1 (HO-1). Reactive Oxygen Species 130-153 heme oxygenase 1 Homo sapiens 281-297 26993595-4 2016 We show that the effects of CO on tumor stroma and reprogramming of macrophages towards the anti-tumoral phenotype is mediated by reactive oxygen species (ROS)-dependent activation of MAPK/Erk1/2-c-myc pathway as well as Notch 1-dependent negative feedback on the metabolic enzyme heme oxygenase-1 (HO-1). Reactive Oxygen Species 130-153 heme oxygenase 1 Homo sapiens 299-303 26993595-4 2016 We show that the effects of CO on tumor stroma and reprogramming of macrophages towards the anti-tumoral phenotype is mediated by reactive oxygen species (ROS)-dependent activation of MAPK/Erk1/2-c-myc pathway as well as Notch 1-dependent negative feedback on the metabolic enzyme heme oxygenase-1 (HO-1). Reactive Oxygen Species 155-158 heme oxygenase 1 Homo sapiens 281-297 26993595-4 2016 We show that the effects of CO on tumor stroma and reprogramming of macrophages towards the anti-tumoral phenotype is mediated by reactive oxygen species (ROS)-dependent activation of MAPK/Erk1/2-c-myc pathway as well as Notch 1-dependent negative feedback on the metabolic enzyme heme oxygenase-1 (HO-1). Reactive Oxygen Species 155-158 heme oxygenase 1 Homo sapiens 299-303 26679389-8 2016 RESULTS: Our results show that monomethyl fumarate induced nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 and concomitant production of the antioxidant enzymes heme oxygenase-1 and NADPH:quinone oxidoreductase-1 in brain endothelial cells. citraconic acid 31-50 heme oxygenase 1 Homo sapiens 182-198 27459794-3 2016 Western blot was used to evaluate the expression of Nrf2, quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (10-1) during sodium arsenite-induced transformation of HBE cells. sodium arsenite 125-140 heme oxygenase 1 Homo sapiens 94-110 27459794-7 2016 CONCLUSION: The transformation of HBE cells induced by chronic exposure to sodium arsenite is mediated by decreased Nrf2 level and its downstream NQO1 and HO-1 protein, which subsequently promote the malignant proliferation. sodium arsenite 75-90 heme oxygenase 1 Homo sapiens 155-159 27097841-3 2016 The results showed a time-dependent HO-1 upregulation in primary cultured ventral mesencephalon neurons and astrocytes treated with the mitochondria complex I inhibitor 1-methyl-4-phenylpyridinium (MPP(+)) or recombinant alpha-synuclein. 1-Methyl-4-phenylpyridinium 169-196 heme oxygenase 1 Homo sapiens 36-40 27097841-3 2016 The results showed a time-dependent HO-1 upregulation in primary cultured ventral mesencephalon neurons and astrocytes treated with the mitochondria complex I inhibitor 1-methyl-4-phenylpyridinium (MPP(+)) or recombinant alpha-synuclein. mangion-purified polysaccharide (Candida albicans) 198-201 heme oxygenase 1 Homo sapiens 36-40 27097841-5 2016 The HO-1 inhibitor zinc protoporphyrin (ZnPP) aggravated MPP(+)- or alpha-synuclein-induced oxidative damage in both astrocytes and neurons, indicating that this HO-1 response was cytoprotective. zinc protoporphyrin 19-38 heme oxygenase 1 Homo sapiens 4-8 27097841-5 2016 The HO-1 inhibitor zinc protoporphyrin (ZnPP) aggravated MPP(+)- or alpha-synuclein-induced oxidative damage in both astrocytes and neurons, indicating that this HO-1 response was cytoprotective. zinc protoporphyrin 19-38 heme oxygenase 1 Homo sapiens 162-166 27097841-5 2016 The HO-1 inhibitor zinc protoporphyrin (ZnPP) aggravated MPP(+)- or alpha-synuclein-induced oxidative damage in both astrocytes and neurons, indicating that this HO-1 response was cytoprotective. zinc protoporphyrin 40-44 heme oxygenase 1 Homo sapiens 4-8 27097841-5 2016 The HO-1 inhibitor zinc protoporphyrin (ZnPP) aggravated MPP(+)- or alpha-synuclein-induced oxidative damage in both astrocytes and neurons, indicating that this HO-1 response was cytoprotective. zinc protoporphyrin 40-44 heme oxygenase 1 Homo sapiens 162-166 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 32-56 heme oxygenase 1 Homo sapiens 17-21 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 32-56 heme oxygenase 1 Homo sapiens 143-147 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 58-64 heme oxygenase 1 Homo sapiens 17-21 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 58-64 heme oxygenase 1 Homo sapiens 143-147 27097841-6 2016 For neurons, the HO-1 activator cobalt protoporphyrin IX (CoPPIX) exerted protective effects against MPP(+) or alpha-synuclein during moderate HO-1 upregulation, but it aggravated damage at the peak of the HO-1 response. cobaltiprotoporphyrin 58-64 heme oxygenase 1 Homo sapiens 143-147 26595750-1 2016 Heme oxygenase (HO)-1 catalyzes the degradation of cytotoxic heme into biliverdin and blocks antitumor immune responses, thus protecting cancer against host defense. Heme 61-65 heme oxygenase 1 Homo sapiens 0-21 26595750-1 2016 Heme oxygenase (HO)-1 catalyzes the degradation of cytotoxic heme into biliverdin and blocks antitumor immune responses, thus protecting cancer against host defense. Biliverdine 71-81 heme oxygenase 1 Homo sapiens 0-21 26566708-1 2016 Heme oxygenase (HO)-1 is upregulated by many stressful stimuli, including arsenic. Arsenic 74-81 heme oxygenase 1 Homo sapiens 0-21 26595750-5 2016 On the basis of this observation we aimed to target HO-1 by systemic as well as tumor-specific zinc protoporphyrin-mediated HO-1 suppression in a syngeneic immunocompetent NB mouse model. zinc protoporphyrin 95-114 heme oxygenase 1 Homo sapiens 124-128 26566708-4 2016 We prospectively investigated the associations between HO-1 (GT)n polymorphism and cancer risk related to arsenic from drinking water. Arsenic 106-113 heme oxygenase 1 Homo sapiens 55-59 26945724-0 2016 Iron depletion in HCT116 cells diminishes the upregulatory effect of phenethyl isothiocyanate on heme oxygenase-1. Iron 0-4 heme oxygenase 1 Homo sapiens 97-113 26566708-4 2016 We prospectively investigated the associations between HO-1 (GT)n polymorphism and cancer risk related to arsenic from drinking water. Water 128-133 heme oxygenase 1 Homo sapiens 55-59 26566708-13 2016 Associations of HO-1 (GT)n polymorphism with cancer risk differs by histological subtype and the polymorphism should be considered a modifier in the risk assessment of arsenic exposure. Arsenic 168-175 heme oxygenase 1 Homo sapiens 16-20 26945724-0 2016 Iron depletion in HCT116 cells diminishes the upregulatory effect of phenethyl isothiocyanate on heme oxygenase-1. phenethyl isothiocyanate 69-93 heme oxygenase 1 Homo sapiens 97-113 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). Iron 51-55 heme oxygenase 1 Homo sapiens 113-129 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). Iron 51-55 heme oxygenase 1 Homo sapiens 131-135 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). phenethyl isothiocyanate 153-177 heme oxygenase 1 Homo sapiens 113-129 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). phenethyl isothiocyanate 153-177 heme oxygenase 1 Homo sapiens 131-135 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). phenethyl isothiocyanate 179-184 heme oxygenase 1 Homo sapiens 113-129 26945724-3 2016 Herein, the objective was to ascertain if adequate iron is needed for enabling HCT116 cells to optimally express heme oxygenase-1 (HO-1) when induced by phenethyl isothiocyanate (PEITC). phenethyl isothiocyanate 179-184 heme oxygenase 1 Homo sapiens 131-135 26945724-7 2016 To elucidate these findings, it was found that PEITC-induced HO-1 upregulation can be inhibited with thiol antioxidants (glutathione and N-acetylcysteine). phenethyl isothiocyanate 47-52 heme oxygenase 1 Homo sapiens 61-65 26945724-7 2016 To elucidate these findings, it was found that PEITC-induced HO-1 upregulation can be inhibited with thiol antioxidants (glutathione and N-acetylcysteine). Sulfhydryl Compounds 101-106 heme oxygenase 1 Homo sapiens 61-65 26945724-7 2016 To elucidate these findings, it was found that PEITC-induced HO-1 upregulation can be inhibited with thiol antioxidants (glutathione and N-acetylcysteine). Glutathione 121-132 heme oxygenase 1 Homo sapiens 61-65 26945724-7 2016 To elucidate these findings, it was found that PEITC-induced HO-1 upregulation can be inhibited with thiol antioxidants (glutathione and N-acetylcysteine). Acetylcysteine 137-153 heme oxygenase 1 Homo sapiens 61-65 26945724-8 2016 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin) and a superoxide scavenger (Tiron) each inhibited PEITC-induced HO-1 upregulation. diphenyleneiodonium 39-58 heme oxygenase 1 Homo sapiens 137-141 26945724-8 2016 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin) and a superoxide scavenger (Tiron) each inhibited PEITC-induced HO-1 upregulation. acetovanillone 63-71 heme oxygenase 1 Homo sapiens 137-141 26945724-8 2016 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin) and a superoxide scavenger (Tiron) each inhibited PEITC-induced HO-1 upregulation. Superoxides 79-89 heme oxygenase 1 Homo sapiens 137-141 26945724-8 2016 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin) and a superoxide scavenger (Tiron) each inhibited PEITC-induced HO-1 upregulation. 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt 101-106 heme oxygenase 1 Homo sapiens 137-141 26945724-8 2016 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin) and a superoxide scavenger (Tiron) each inhibited PEITC-induced HO-1 upregulation. phenethyl isothiocyanate 123-128 heme oxygenase 1 Homo sapiens 137-141 26945724-10 2016 Collectively, the results imply that the HO-1 upregulation by PEITC involves an iron-dependent, oxidant signaling pathway. Iron 80-84 heme oxygenase 1 Homo sapiens 41-45 26945724-11 2016 Therefore, it is concluded that ample iron is required to enable PEITC to fully upregulate HO-1 expression in HCT116 cells. Iron 38-42 heme oxygenase 1 Homo sapiens 91-95 26792743-6 2016 Engagement of the hIL-10 receptor (hIL-10R) by cmvIL-10 led to upregulation of heme oxygenase 1 (HO-1), an enzyme linked with suppression of inflammatory responses, and this upregulation was required for cmvIL-10-mediated upregulation of hIL-10. cmvil-10 47-55 heme oxygenase 1 Homo sapiens 79-95 27071063-3 2016 We found that activation of NRF2 with sulforaphane (SFN) in erythroid progenitors significantly increased the expression of NRF2 targets HMOX1, NQO1, and HBG1 (subunit of fetal hemoglobin) in a dose-dependent manner. sulforaphane 38-50 heme oxygenase 1 Homo sapiens 137-142 27071063-3 2016 We found that activation of NRF2 with sulforaphane (SFN) in erythroid progenitors significantly increased the expression of NRF2 targets HMOX1, NQO1, and HBG1 (subunit of fetal hemoglobin) in a dose-dependent manner. sulforaphane 52-55 heme oxygenase 1 Homo sapiens 137-142 27071063-8 2016 We observed an increase in the mean relative whole blood mRNA levels for the NRF2 target HMOX1 (p = 0.02) on the last day of BSH treatment, compared to pre-treatment. benzenesulfohydrazide 125-128 heme oxygenase 1 Homo sapiens 89-94 27058954-8 2016 In addition to neuroglioma cells, we also found Abeta-induced epigenetic regulation of HMOX1 in human T lymphocyte Jurkat cells. UNII-042A8N37WH 48-53 heme oxygenase 1 Homo sapiens 87-92 26801320-0 2016 Inhibition of heme oxygenase-1 enhances the chemosensitivity of laryngeal squamous cell cancer Hep-2 cells to cisplatin. Cisplatin 110-119 heme oxygenase 1 Homo sapiens 14-30 26801320-2 2016 In this research we proved that cisplatin induced cell injuries and heme oxygenase-1 (HO-1) expression in laryngeal squamous cell cancer Hep-2 cells through ROS generation. Cisplatin 32-41 heme oxygenase 1 Homo sapiens 68-84 26801320-2 2016 In this research we proved that cisplatin induced cell injuries and heme oxygenase-1 (HO-1) expression in laryngeal squamous cell cancer Hep-2 cells through ROS generation. Cisplatin 32-41 heme oxygenase 1 Homo sapiens 86-90 26801320-2 2016 In this research we proved that cisplatin induced cell injuries and heme oxygenase-1 (HO-1) expression in laryngeal squamous cell cancer Hep-2 cells through ROS generation. ros 157-160 heme oxygenase 1 Homo sapiens 68-84 26801320-2 2016 In this research we proved that cisplatin induced cell injuries and heme oxygenase-1 (HO-1) expression in laryngeal squamous cell cancer Hep-2 cells through ROS generation. ros 157-160 heme oxygenase 1 Homo sapiens 86-90 26801320-3 2016 The induction of HO-1 clearly protected Hep-2 cells from cisplatin-induced cell death and ROS reaction, and the inhibitor of HO-1 enhanced the cell death and ROS generation induced by cisplatin. Cisplatin 57-66 heme oxygenase 1 Homo sapiens 17-21 26801320-3 2016 The induction of HO-1 clearly protected Hep-2 cells from cisplatin-induced cell death and ROS reaction, and the inhibitor of HO-1 enhanced the cell death and ROS generation induced by cisplatin. ros 90-93 heme oxygenase 1 Homo sapiens 17-21 26801320-3 2016 The induction of HO-1 clearly protected Hep-2 cells from cisplatin-induced cell death and ROS reaction, and the inhibitor of HO-1 enhanced the cell death and ROS generation induced by cisplatin. ros 158-161 heme oxygenase 1 Homo sapiens 125-129 26801320-3 2016 The induction of HO-1 clearly protected Hep-2 cells from cisplatin-induced cell death and ROS reaction, and the inhibitor of HO-1 enhanced the cell death and ROS generation induced by cisplatin. Cisplatin 184-193 heme oxygenase 1 Homo sapiens 125-129 26801320-4 2016 Furthermore, the HO-1 expression induced by cisplatin was strongly inhibited by the knockdown of nuclear factor-erythroid-2-related factor-2 (Nrf-2), and the oxidative damages induced by cisplatin were significantly enhanced. Cisplatin 44-53 heme oxygenase 1 Homo sapiens 17-21 26801320-4 2016 Furthermore, the HO-1 expression induced by cisplatin was strongly inhibited by the knockdown of nuclear factor-erythroid-2-related factor-2 (Nrf-2), and the oxidative damages induced by cisplatin were significantly enhanced. Cisplatin 187-196 heme oxygenase 1 Homo sapiens 17-21 26801320-5 2016 Therefore, it may be concluded that the inhibition of HO-1 or the knockdown of Nrf-2 significantly enhanced cisplatin"s anticancer effects on Hep-2 cells. Cisplatin 108-117 heme oxygenase 1 Homo sapiens 54-58 26801320-6 2016 In clinic, with the overexpression of HO-1 in laryngeal squamous cancer tissues, the combination of cisplatin with the inhibitor of HO-1 or Nrf-2 siRNA may act as a new method to the treatment of laryngeal squamous cancer. Cisplatin 100-109 heme oxygenase 1 Homo sapiens 38-42 26801320-6 2016 In clinic, with the overexpression of HO-1 in laryngeal squamous cancer tissues, the combination of cisplatin with the inhibitor of HO-1 or Nrf-2 siRNA may act as a new method to the treatment of laryngeal squamous cancer. Cisplatin 100-109 heme oxygenase 1 Homo sapiens 132-136 26780849-15 2016 Knockdown of HO-1 promoted apoptosis through activation of a ROS-mediated caspase apoptosis pathway. Reactive Oxygen Species 61-64 heme oxygenase 1 Homo sapiens 13-17 27023064-0 2016 Role of Nrf2, HO-1 and GSH in Neuroblastoma Cell Resistance to Bortezomib. Bortezomib 63-73 heme oxygenase 1 Homo sapiens 14-18 27023064-4 2016 BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of gamma-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. Bortezomib 0-3 heme oxygenase 1 Homo sapiens 111-127 27023064-4 2016 BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of gamma-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. Bortezomib 0-3 heme oxygenase 1 Homo sapiens 129-133 27023064-4 2016 BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of gamma-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. Cysteine 176-184 heme oxygenase 1 Homo sapiens 111-127 27023064-4 2016 BTZ treatment favours the binding of Nrf2 to the ARE sequences in the promoter regions of target genes such as heme oxygenase 1 (HO-1), the modulatory subunit of gamma-glutamylcysteine ligase (GCLM) and the transporter for cysteine (x-CT), enabling their transcription. Cysteine 176-184 heme oxygenase 1 Homo sapiens 129-133 27023064-6 2016 The up-regulation of Nrf2 target genes is responsible for cell resistance since HO-1 silencing and GSH depletion synergistically decrease BTZ-treated cell viability. Bortezomib 138-141 heme oxygenase 1 Homo sapiens 80-84 27023064-7 2016 Moreover, cell exposure to all-trans-Retinoic acid (ATRA, 3 muM) reduces the binding of Nrf2 to the ARE sequences, decreases HO-1 induction and lowers GSH level increasing the efficacy of bortezomib. Tretinoin 27-50 heme oxygenase 1 Homo sapiens 125-129 27023064-7 2016 Moreover, cell exposure to all-trans-Retinoic acid (ATRA, 3 muM) reduces the binding of Nrf2 to the ARE sequences, decreases HO-1 induction and lowers GSH level increasing the efficacy of bortezomib. Tretinoin 52-56 heme oxygenase 1 Homo sapiens 125-129 27023064-7 2016 Moreover, cell exposure to all-trans-Retinoic acid (ATRA, 3 muM) reduces the binding of Nrf2 to the ARE sequences, decreases HO-1 induction and lowers GSH level increasing the efficacy of bortezomib. Bortezomib 188-198 heme oxygenase 1 Homo sapiens 125-129 27023064-8 2016 These data suggest the role of Nrf2, HO-1 and GSH as molecular targets to improve the efficacy of low doses of bortezomib in the treatment of malignant neuroblastoma. Bortezomib 111-121 heme oxygenase 1 Homo sapiens 37-41 26792743-6 2016 Engagement of the hIL-10 receptor (hIL-10R) by cmvIL-10 led to upregulation of heme oxygenase 1 (HO-1), an enzyme linked with suppression of inflammatory responses, and this upregulation was required for cmvIL-10-mediated upregulation of hIL-10. cmvil-10 47-55 heme oxygenase 1 Homo sapiens 97-101 26792743-6 2016 Engagement of the hIL-10 receptor (hIL-10R) by cmvIL-10 led to upregulation of heme oxygenase 1 (HO-1), an enzyme linked with suppression of inflammatory responses, and this upregulation was required for cmvIL-10-mediated upregulation of hIL-10. cmvil-10 204-212 heme oxygenase 1 Homo sapiens 79-95 26792743-6 2016 Engagement of the hIL-10 receptor (hIL-10R) by cmvIL-10 led to upregulation of heme oxygenase 1 (HO-1), an enzyme linked with suppression of inflammatory responses, and this upregulation was required for cmvIL-10-mediated upregulation of hIL-10. cmvil-10 204-212 heme oxygenase 1 Homo sapiens 97-101 26801686-0 2016 Activation of the MAPK/Akt/Nrf2-Egr1/HO-1-GCLc axis protects MG-63 osteosarcoma cells against 15d-PGJ2-mediated cell death. 9-deoxy-delta-9-prostaglandin D2 98-102 heme oxygenase 1 Homo sapiens 37-41 26876575-0 2016 MicroRNA-218 promotes high glucose-induced apoptosis in podocytes by targeting heme oxygenase-1. Glucose 27-34 heme oxygenase 1 Homo sapiens 79-95 27023525-18 2016 CORM-2 decreased NO and increased HO-1 expression and CO and COHb content, downregulated HIF-1alpha, as well as WRS-elevated COX-2 and iNOS mRNAs. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 34-38 26801686-5 2016 This pathway induced nuclear expression of Nrf2 and Egr1, and increased transcription of haem oxygenase-1 (HO-1) and the catalytic subunit of glutamate cysteine ligase (GCLc), catalysing the first step in GSH synthesis. Glutathione 205-208 heme oxygenase 1 Homo sapiens 89-105 26801686-5 2016 This pathway induced nuclear expression of Nrf2 and Egr1, and increased transcription of haem oxygenase-1 (HO-1) and the catalytic subunit of glutamate cysteine ligase (GCLc), catalysing the first step in GSH synthesis. Glutathione 205-208 heme oxygenase 1 Homo sapiens 107-111 26801686-6 2016 Silencing of Nrf2, Egr1 and HO-1 significantly elevated 15d-PGJ2-mediated reduction of cellular metabolic activity. 15-deoxy-delta(12,14)-prostaglandin J2 56-64 heme oxygenase 1 Homo sapiens 28-32 26801686-7 2016 Activation of cell survival genes including HO-1 and GCLc inhibited 15d-PGJ2-induced cleavage of pro-caspase-3 and PARP. 9-deoxy-delta-9-prostaglandin D2 72-76 heme oxygenase 1 Homo sapiens 44-48 26801686-12 2016 Pharmacological or genetic interference of the pro-survival pathway, the p38 MAPK/Akt/Nrf2-Egr1/HO-1-GCLc axis, sensitizes MG-63 cells towards 15d-PGJ2-mediated apoptosis. 9-deoxy-delta-9-prostaglandin D2 147-151 heme oxygenase 1 Homo sapiens 96-105 26725491-0 2016 Heme Oxygenase-1/Carbon Monoxide System and Embryonic Stem Cell Differentiation and Maturation into Cardiomyocytes. Carbon Monoxide 17-32 heme oxygenase 1 Homo sapiens 0-16 26725491-3 2016 In this study, we tested whether the redox-sensitive heme oxygenase-1/carbon monoxide (HO-1/CO) system, operating through mitochondrial biogenesis, acts as a mechanism for ES cell differentiation and cardiomyocyte maturation. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 53-69 26725491-3 2016 In this study, we tested whether the redox-sensitive heme oxygenase-1/carbon monoxide (HO-1/CO) system, operating through mitochondrial biogenesis, acts as a mechanism for ES cell differentiation and cardiomyocyte maturation. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 87-94 26898422-0 2016 Heme oxygenase-1 contributes to imatinib resistance by promoting autophagy in chronic myeloid leukemia through disrupting the mTOR signaling pathway. Imatinib Mesylate 32-40 heme oxygenase 1 Homo sapiens 0-16 26700310-0 2016 Potential crosstalk of the interleukin-6-heme oxygenase-1-dependent mechanism involved in resistance to lenalidomide in multiple myeloma cells. Lenalidomide 104-116 heme oxygenase 1 Homo sapiens 41-57 26700310-7 2016 In contrast, without IL-6 coculture, enhanced HO-1 expression in U266, RPMI8226 and bone marrow CD138(+) cells from MM patients significantly inreased IL-6 mRNA expression levels and facilitated autocrine IL-6 production. rpmi8226 71-79 heme oxygenase 1 Homo sapiens 46-50 26898422-1 2016 Heme oxygenase-1 (HO-1) has been verified to play an important role in imatinib (IM)-resistant chronic myeloid leukemia (CML) cells, but the mechanism remains unclear. Imatinib Mesylate 71-79 heme oxygenase 1 Homo sapiens 0-16 26700310-9 2016 Reduced HO-1 expression sensitized MM cells to lenalidomide. Lenalidomide 47-59 heme oxygenase 1 Homo sapiens 8-12 26898422-1 2016 Heme oxygenase-1 (HO-1) has been verified to play an important role in imatinib (IM)-resistant chronic myeloid leukemia (CML) cells, but the mechanism remains unclear. Imatinib Mesylate 71-79 heme oxygenase 1 Homo sapiens 18-22 27051648-6 2016 In the present study, the protective effects of hyperoside against hydrogen peroxide-induced oxidative stress in human lens epithelial cells, HLE-B3, were investigated in terms of HO-1 induction. hyperoside 48-58 heme oxygenase 1 Homo sapiens 180-184 26698668-4 2016 Affymetrix array profiling and subsequent qPCR/protein validation revealed that induction of select Nrf2 target genes, HO-1 and NQO1, was significantly attenuated in cells adapted to 5% O2, despite nuclear accumulation and DNA binding of Nrf2. Oxygen 186-188 heme oxygenase 1 Homo sapiens 119-123 26698668-5 2016 Diminished HO-1 induction under 5% O2 was stimulus independent and reversible upon re-adaptation to air or silencing of the Nrf2 repressor Bach1, notably elevated under 5% O2. Oxygen 35-37 heme oxygenase 1 Homo sapiens 11-15 26698668-5 2016 Diminished HO-1 induction under 5% O2 was stimulus independent and reversible upon re-adaptation to air or silencing of the Nrf2 repressor Bach1, notably elevated under 5% O2. Oxygen 172-174 heme oxygenase 1 Homo sapiens 11-15 27051648-11 2016 RESULTS: Hyperoside increased both the mRNA and protein expression of HO-1 in a time- and dose-dependent manner. hyperoside 9-19 heme oxygenase 1 Homo sapiens 70-74 27051648-14 2016 CONCLUSIONS: Hyperoside is an effective compound to protect cells against oxidative stress via HO-1 induction. hyperoside 13-23 heme oxygenase 1 Homo sapiens 95-99 27263286-3 2016 RESULTS: 10 mumol/L arsenic trioxide can decreased the cell viability, while cell apoptosis rate, ROS level, HO-1 and Nrf2 protein expression was increased (P < 0.05). Arsenic Trioxide 20-36 heme oxygenase 1 Homo sapiens 109-113 26681403-11 2016 In contrast, Nrf2 and HO-1 expression were markedly increased (P = 0.0001, P = 0.0049, respectively) after smokers took melatonin orally. Melatonin 120-129 heme oxygenase 1 Homo sapiens 22-26 27263286-4 2016 When compared with arsenic trioxide alone, co-treatment of arsenic trioxide with aspirin in different concentration (0, 0.1, 1.0, 2.5, 5.0 mmol/L) exhibited dual effects in intracellular ROS level, HO-1 and Nrf2 expression. Arsenic Trioxide 59-75 heme oxygenase 1 Homo sapiens 198-202 27263286-7 2016 On the contrary, 5 mmol/L aspirin could increase the sensitivity of HepG2 to arsenic trioxide through enhancing the arsenic trioxide-induced apoptosis by ROS accumulation resulting in inhibiting the Nrf2-HO-1 pathway. Arsenic Trioxide 116-132 heme oxygenase 1 Homo sapiens 204-208 27263286-4 2016 When compared with arsenic trioxide alone, co-treatment of arsenic trioxide with aspirin in different concentration (0, 0.1, 1.0, 2.5, 5.0 mmol/L) exhibited dual effects in intracellular ROS level, HO-1 and Nrf2 expression. Aspirin 81-88 heme oxygenase 1 Homo sapiens 198-202 27263286-5 2016 Specifically, with the increasing of aspirin concentrations, the level of ROS induced by arsenic trioxide showed a rising trend after the first reduction, whereas, HO-1 and Nrf2 protein expression were decreased at first and then increased. Aspirin 37-44 heme oxygenase 1 Homo sapiens 164-168 27263286-6 2016 CONCLUSION: Low concentration, less than 2.5 mmol/L, of aspirin may reduce the ROS accumulation through activating of Nrf2-HO-1 pathway, therefore decreasing the apoptotic cell death induced by arsenic trioxide. Aspirin 56-63 heme oxygenase 1 Homo sapiens 123-127 27263286-7 2016 On the contrary, 5 mmol/L aspirin could increase the sensitivity of HepG2 to arsenic trioxide through enhancing the arsenic trioxide-induced apoptosis by ROS accumulation resulting in inhibiting the Nrf2-HO-1 pathway. Aspirin 26-33 heme oxygenase 1 Homo sapiens 204-208 27263286-7 2016 On the contrary, 5 mmol/L aspirin could increase the sensitivity of HepG2 to arsenic trioxide through enhancing the arsenic trioxide-induced apoptosis by ROS accumulation resulting in inhibiting the Nrf2-HO-1 pathway. Arsenic Trioxide 77-93 heme oxygenase 1 Homo sapiens 204-208 27033569-10 2016 The HO-1 mRNA level before the stimulation was 1.000, and 17.264+-4.275 after the stimulation of 1 ng/ml IL-17A (U=0, P<0.05), 19.128+-4.605 after the stimulation of 10 ng/ml IL-17A (U=0, P<0.05), but was significantly suppressed after stimulation with glucocorticoids (dexamethasone, DEX). Dexamethasone 276-289 heme oxygenase 1 Homo sapiens 4-8 27033569-10 2016 The HO-1 mRNA level before the stimulation was 1.000, and 17.264+-4.275 after the stimulation of 1 ng/ml IL-17A (U=0, P<0.05), 19.128+-4.605 after the stimulation of 10 ng/ml IL-17A (U=0, P<0.05), but was significantly suppressed after stimulation with glucocorticoids (dexamethasone, DEX). Dextromethorphan 291-294 heme oxygenase 1 Homo sapiens 4-8 26869854-10 2016 Noteworthy, the impaired TCA cycle in HT-1080 cells were associated with high mTORC1 activity, negligible protein level and activity of mTORC2, high expression of interleukin-1beta, interleukin-6 and heme oxygenase-1 which may contribute to the compensatory mechanism of TCA deficiency. Trichloroacetic Acid 25-28 heme oxygenase 1 Homo sapiens 200-216 30263273-7 2016 6-shogaol exerts an indirect cellular anti-oxidant activity based on up-regulation of GCS and HO-1 via a JNK-mediated Nrf2 signaling pathway. shogaol 0-9 heme oxygenase 1 Homo sapiens 94-98 26721585-1 2016 Carbon monoxide (CO) is generated by heme oxygenase-1 (HO-1) and displays important signaling, anti-apoptotic and anti-inflammatory activities, indicating that pharmacological agents mimicking its action may have therapeutic benefit. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 37-53 26721585-1 2016 Carbon monoxide (CO) is generated by heme oxygenase-1 (HO-1) and displays important signaling, anti-apoptotic and anti-inflammatory activities, indicating that pharmacological agents mimicking its action may have therapeutic benefit. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 55-59 26721585-1 2016 Carbon monoxide (CO) is generated by heme oxygenase-1 (HO-1) and displays important signaling, anti-apoptotic and anti-inflammatory activities, indicating that pharmacological agents mimicking its action may have therapeutic benefit. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 37-53 26721585-1 2016 Carbon monoxide (CO) is generated by heme oxygenase-1 (HO-1) and displays important signaling, anti-apoptotic and anti-inflammatory activities, indicating that pharmacological agents mimicking its action may have therapeutic benefit. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 55-59 26483091-1 2016 BACKGROUND: Heme-oxygenase 1 (HO-1), an inducible heme-degrading enzyme, has antiatherogenic effects through its enzymatic end products. Heme 50-54 heme oxygenase 1 Homo sapiens 12-28 26866373-10 2016 EGCG down-regulated NLRP3 expression, which was blocked by ZnPP, indicating that HO-1 links EGCG with NLRP3. epigallocatechin gallate 0-4 heme oxygenase 1 Homo sapiens 81-85 26866373-10 2016 EGCG down-regulated NLRP3 expression, which was blocked by ZnPP, indicating that HO-1 links EGCG with NLRP3. zinc protoporphyrin 59-63 heme oxygenase 1 Homo sapiens 81-85 26866373-10 2016 EGCG down-regulated NLRP3 expression, which was blocked by ZnPP, indicating that HO-1 links EGCG with NLRP3. epigallocatechin gallate 92-96 heme oxygenase 1 Homo sapiens 81-85 26866373-11 2016 Therefore, EGCG, via up-regulation of HO-1, protects against CIN by amelioration of oxidative stress and inflammation. epigallocatechin gallate 11-15 heme oxygenase 1 Homo sapiens 38-42 26723515-6 2016 Furthermore, eriodictyol was found to up-regulate the expression of Nrf2/HO-1 and inhibited CP-induced NF-kappaB activation in kidney tissues. eriodictyol 13-24 heme oxygenase 1 Homo sapiens 73-77 26845693-0 2016 Modification of Caffeic Acid with Pyrrolidine Enhances Antioxidant Ability by Activating AKT/HO-1 Pathway in Heart. caffeic acid 16-28 heme oxygenase 1 Homo sapiens 93-97 26845693-0 2016 Modification of Caffeic Acid with Pyrrolidine Enhances Antioxidant Ability by Activating AKT/HO-1 Pathway in Heart. pyrrolidine 34-45 heme oxygenase 1 Homo sapiens 93-97 25575725-6 2016 Studies of human umbilical vein endothelial cells (HUVEC) and arterial endothelial cells (HAEC) showed that therapeutically relevant concentrations of MTX phosphorylate AMPKalpha(Thr172), and induce cytoprotective genes including manganese superoxide dismutase (MnSOD) and haem oxygenase-1 (HO-1). Methotrexate 151-154 heme oxygenase 1 Homo sapiens 273-289 25575725-6 2016 Studies of human umbilical vein endothelial cells (HUVEC) and arterial endothelial cells (HAEC) showed that therapeutically relevant concentrations of MTX phosphorylate AMPKalpha(Thr172), and induce cytoprotective genes including manganese superoxide dismutase (MnSOD) and haem oxygenase-1 (HO-1). Methotrexate 151-154 heme oxygenase 1 Homo sapiens 291-295 25575725-9 2016 Mechanistically, MTX treatment led to cyclic AMP response element-binding protein (CREB)(Ser133) phosphorylation, while AMPK depletion attenuated this response and the induction of MnSOD and HO-1. Methotrexate 17-20 heme oxygenase 1 Homo sapiens 191-195 26866373-0 2016 Targeting HO-1 by Epigallocatechin-3-Gallate Reduces Contrast-Induced Renal Injury via Anti-Oxidative Stress and Anti-Inflammation Pathways. epigallocatechin gallate 18-44 heme oxygenase 1 Homo sapiens 10-14 26866373-5 2016 Because EGCG is a potent inducer of the antioxidant heme oxygenase-1 (HO-1), we studied HO-1 signaling in CIN. epigallocatechin gallate 8-12 heme oxygenase 1 Homo sapiens 52-68 26866373-5 2016 Because EGCG is a potent inducer of the antioxidant heme oxygenase-1 (HO-1), we studied HO-1 signaling in CIN. epigallocatechin gallate 8-12 heme oxygenase 1 Homo sapiens 70-74 26866373-5 2016 Because EGCG is a potent inducer of the antioxidant heme oxygenase-1 (HO-1), we studied HO-1 signaling in CIN. epigallocatechin gallate 8-12 heme oxygenase 1 Homo sapiens 88-92 26866373-6 2016 HO-1 levels were increased in CIN; treatment with EGCG further increased HO-1 levels, accompanied by an increase in Nrf2, a regulator of antioxidant proteins. epigallocatechin gallate 50-54 heme oxygenase 1 Homo sapiens 0-4 26866373-6 2016 HO-1 levels were increased in CIN; treatment with EGCG further increased HO-1 levels, accompanied by an increase in Nrf2, a regulator of antioxidant proteins. epigallocatechin gallate 50-54 heme oxygenase 1 Homo sapiens 73-77 26866373-7 2016 Interestingly, blockade of HO-1 with protoporphyrin IX zinc(II) (ZnPP) prevented the protective effect of EGCG on CIN. protoporphyrin ix zinc(ii) 37-63 heme oxygenase 1 Homo sapiens 27-31 26866373-7 2016 Interestingly, blockade of HO-1 with protoporphyrin IX zinc(II) (ZnPP) prevented the protective effect of EGCG on CIN. zinc protoporphyrin 65-69 heme oxygenase 1 Homo sapiens 27-31 26866373-7 2016 Interestingly, blockade of HO-1 with protoporphyrin IX zinc(II) (ZnPP) prevented the protective effect of EGCG on CIN. epigallocatechin gallate 106-110 heme oxygenase 1 Homo sapiens 27-31 26866373-8 2016 ZnPP also blocked the ability of EGCG to increase the activity of an antioxidant (superoxide dismutase), and decrease markers of oxidative stress (myeloperoxidase and malondialdehyde) and inflammation (myeloperoxidase and IL-1beta), indicating that HO-1 is the upstream molecule that regulates the EGCG-mediated protection. zinc protoporphyrin 0-4 heme oxygenase 1 Homo sapiens 249-253 26866373-8 2016 ZnPP also blocked the ability of EGCG to increase the activity of an antioxidant (superoxide dismutase), and decrease markers of oxidative stress (myeloperoxidase and malondialdehyde) and inflammation (myeloperoxidase and IL-1beta), indicating that HO-1 is the upstream molecule that regulates the EGCG-mediated protection. epigallocatechin gallate 33-37 heme oxygenase 1 Homo sapiens 249-253 26483091-1 2016 BACKGROUND: Heme-oxygenase 1 (HO-1), an inducible heme-degrading enzyme, has antiatherogenic effects through its enzymatic end products. Heme 50-54 heme oxygenase 1 Homo sapiens 30-34 26483091-2 2016 HO-1 gene expression is modulated by a guanidine thymidine dinucleotide ([GT]n) repeat polymorphism in the promoter region. guanidine thymidine dinucleotide 39-71 heme oxygenase 1 Homo sapiens 0-4 26689473-6 2016 The 27-OH-exerted induction of Nrf2 and subsequently of the target genes, HO-1 and NQO-1, was proved to be: (i) dependent upon the activation of ERK and Akt pathways, (ii) directly responsible for the quenching of intracellular oxidative stress and by this way (iii) ultimately responsible for the observed oxysterol-induced pro-survival response. 27-hydroxycholesterol 4-9 heme oxygenase 1 Homo sapiens 74-78 26467701-8 2016 DHT induced a significant increase in expression of the androgen-responsive genes FKBP5, HMOX1, FBXO32, VEGFA, WNT5A, and KLK3 only in AR-positive cells in vitro. Dihydrotestosterone 0-3 heme oxygenase 1 Homo sapiens 89-94 26721592-10 2016 CA also reversed the APAP-induced decreased mRNA and protein expression of heme oxygenase 1 (HO-1) and NAD(P)H: quinone oxidoreductase 1(NQO1). Acetaminophen 21-25 heme oxygenase 1 Homo sapiens 75-91 26726846-9 2016 Suppression of HO-1 by Zn-protoporphyrin (ZnPP) enhanced cytotoxicity to GEM in CCA cells, validated in CCA xenografts. zn-protoporphyrin 23-40 heme oxygenase 1 Homo sapiens 15-19 26726846-9 2016 Suppression of HO-1 by Zn-protoporphyrin (ZnPP) enhanced cytotoxicity to GEM in CCA cells, validated in CCA xenografts. Zinc protoporphyrin 42-46 heme oxygenase 1 Homo sapiens 15-19 26698667-9 2016 On the other hand, induction of the Nrf2-regulated enzyme heme oxygenase 1 (HO-1) by cardamonin was diminished in Se-replete compared to Se-deficient cells. cardamonin 85-95 heme oxygenase 1 Homo sapiens 58-74 26698667-9 2016 On the other hand, induction of the Nrf2-regulated enzyme heme oxygenase 1 (HO-1) by cardamonin was diminished in Se-replete compared to Se-deficient cells. Selenium 114-116 heme oxygenase 1 Homo sapiens 58-74 26698667-9 2016 On the other hand, induction of the Nrf2-regulated enzyme heme oxygenase 1 (HO-1) by cardamonin was diminished in Se-replete compared to Se-deficient cells. Selenium 137-139 heme oxygenase 1 Homo sapiens 58-74 26721592-11 2016 In addition, HO-1 inhibitor zinc protoporphyrin (ZnPP) and NQO1 inhibitor diminutol (Dim) reduced the protection of CA against APAP-induced hepatotoxicity. zinc protoporphyrin 28-47 heme oxygenase 1 Homo sapiens 13-17 26689473-6 2016 The 27-OH-exerted induction of Nrf2 and subsequently of the target genes, HO-1 and NQO-1, was proved to be: (i) dependent upon the activation of ERK and Akt pathways, (ii) directly responsible for the quenching of intracellular oxidative stress and by this way (iii) ultimately responsible for the observed oxysterol-induced pro-survival response. Oxysterols 307-316 heme oxygenase 1 Homo sapiens 74-78 26721592-11 2016 In addition, HO-1 inhibitor zinc protoporphyrin (ZnPP) and NQO1 inhibitor diminutol (Dim) reduced the protection of CA against APAP-induced hepatotoxicity. zinc protoporphyrin 49-53 heme oxygenase 1 Homo sapiens 13-17 26721592-11 2016 In addition, HO-1 inhibitor zinc protoporphyrin (ZnPP) and NQO1 inhibitor diminutol (Dim) reduced the protection of CA against APAP-induced hepatotoxicity. Acetaminophen 127-131 heme oxygenase 1 Homo sapiens 13-17 26618301-9 2016 The antioxidant response genes (Nqo1 and Hmox1) were moderately associated with polyaromatic hydrocarbons (PAHs) and showed a good correlation (r-Pearson of >0.7) with metals linked to vehicle-related emissions (i.e. Cu, Zn and Sb). polyaromatic hydrocarbons 80-105 heme oxygenase 1 Homo sapiens 41-46 26721592-15 2016 In conclusion, we demonstrated that CA prevented APAP-induced hepatotoxicity by decreasing Keap1 expression, inhibiting binding of Keap1 to Nrf2, and thus activating Nrf2 and leading to increased expression of antioxidative signals including HO-1 and NQO1. Acetaminophen 49-53 heme oxygenase 1 Homo sapiens 242-246 26691217-2 2016 The aim of this study was to assess the effects of CUGBP2-mediated post-transcriptional regulation of COX-2 and HO-1 in pancreatic cancer cells in regard of response to gemcitabine (GEM) treatment. gemcitabine 169-180 heme oxygenase 1 Homo sapiens 112-116 26691217-2 2016 The aim of this study was to assess the effects of CUGBP2-mediated post-transcriptional regulation of COX-2 and HO-1 in pancreatic cancer cells in regard of response to gemcitabine (GEM) treatment. gemcitabine 182-185 heme oxygenase 1 Homo sapiens 112-116 26691217-9 2016 Induction of CUGBP2 expression by curcumin resulted in the downregulation of HO-1 and COX-2 and strongly sensitized tumor cells to GEM treatment. Curcumin 34-42 heme oxygenase 1 Homo sapiens 77-81 27186034-2 2016 Activity of heme oxygenase-1 is regulated by a guanine-thymine dinucleotide length polymorphism in the heme oxygenase-1 gene promoter. guanine-thymine dinucleotide 47-75 heme oxygenase 1 Homo sapiens 12-28 27186034-2 2016 Activity of heme oxygenase-1 is regulated by a guanine-thymine dinucleotide length polymorphism in the heme oxygenase-1 gene promoter. guanine-thymine dinucleotide 47-75 heme oxygenase 1 Homo sapiens 103-119 26827637-0 2016 Sulforaphane exerts its anti-inflammatory effect against amyloid-beta peptide via STAT-1 dephosphorylation and activation of Nrf2/HO-1 cascade in human THP-1 macrophages. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 130-134 26827637-5 2016 Sulforaphane also increased nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, which was followed by upregulation of heme-oxygenase 1 (HO-1). sulforaphane 0-12 heme oxygenase 1 Homo sapiens 140-156 26827637-5 2016 Sulforaphane also increased nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation, which was followed by upregulation of heme-oxygenase 1 (HO-1). sulforaphane 0-12 heme oxygenase 1 Homo sapiens 158-162 26827637-6 2016 The anti-inflammatory effect of sulforaphane on Abeta1-42-induced IL-1beta production was diminished by small interfering RNA-mediated knockdown of Nrf2 or HO-1. sulforaphane 32-44 heme oxygenase 1 Homo sapiens 156-160 26827637-9 2016 These results indicate that signal transducer and activator of transcription-1 dephosphorylation, HO-1 and its upstream effector, Nrf2, play a pivotal role in triggering an anti-inflammatory signaling cascade of sulforaphane that results in decreases of IL-1beta release and microRNA-146a production in Abeta1-42-stimulated human microglia-like cells. sulforaphane 212-224 heme oxygenase 1 Homo sapiens 98-102 26618301-9 2016 The antioxidant response genes (Nqo1 and Hmox1) were moderately associated with polyaromatic hydrocarbons (PAHs) and showed a good correlation (r-Pearson of >0.7) with metals linked to vehicle-related emissions (i.e. Cu, Zn and Sb). pahs 107-111 heme oxygenase 1 Homo sapiens 41-46 26618301-9 2016 The antioxidant response genes (Nqo1 and Hmox1) were moderately associated with polyaromatic hydrocarbons (PAHs) and showed a good correlation (r-Pearson of >0.7) with metals linked to vehicle-related emissions (i.e. Cu, Zn and Sb). Copper 220-222 heme oxygenase 1 Homo sapiens 41-46 26618301-9 2016 The antioxidant response genes (Nqo1 and Hmox1) were moderately associated with polyaromatic hydrocarbons (PAHs) and showed a good correlation (r-Pearson of >0.7) with metals linked to vehicle-related emissions (i.e. Cu, Zn and Sb). Zinc 224-226 heme oxygenase 1 Homo sapiens 41-46 26618301-9 2016 The antioxidant response genes (Nqo1 and Hmox1) were moderately associated with polyaromatic hydrocarbons (PAHs) and showed a good correlation (r-Pearson of >0.7) with metals linked to vehicle-related emissions (i.e. Cu, Zn and Sb). Antimony 231-233 heme oxygenase 1 Homo sapiens 41-46 26730678-2 2016 We synthesized a series of cobalt-based hybrid molecules (HYCOs) that combine an Nrf2 inducer with a releaser of carbon monoxide (CO), an anti-inflammatory product of HO-1. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 167-171 26730678-2 2016 We synthesized a series of cobalt-based hybrid molecules (HYCOs) that combine an Nrf2 inducer with a releaser of carbon monoxide (CO), an anti-inflammatory product of HO-1. Carbon Monoxide 60-62 heme oxygenase 1 Homo sapiens 167-171 26730678-0 2016 Diverse Nrf2 Activators Coordinated to Cobalt Carbonyls Induce Heme Oxygenase-1 and Release Carbon Monoxide in Vitro and in Vivo. cobalt carbonyls 39-55 heme oxygenase 1 Homo sapiens 63-79 26655464-0 2016 Pinocembrin attenuates MPP(+)-induced neurotoxicity by the induction of heme oxygenase-1 through ERK1/2 pathway. pinocembrin 0-11 heme oxygenase 1 Homo sapiens 72-88 26730678-2 2016 We synthesized a series of cobalt-based hybrid molecules (HYCOs) that combine an Nrf2 inducer with a releaser of carbon monoxide (CO), an anti-inflammatory product of HO-1. Cobalt 27-33 heme oxygenase 1 Homo sapiens 167-171 26822586-0 2016 Blocking heme oxygenase-1 by zinc protoporphyrin reduces tumor hypoxia-mediated VEGF release and inhibits tumor angiogenesis as a potential therapeutic agent against colorectal cancer. zinc protoporphyrin 29-48 heme oxygenase 1 Homo sapiens 9-25 26822586-3 2016 Zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, is potential to inhibit tumor proliferation and progression. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 30-46 26822586-3 2016 Zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, is potential to inhibit tumor proliferation and progression. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 48-52 26822586-3 2016 Zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, is potential to inhibit tumor proliferation and progression. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 30-46 26822586-3 2016 Zinc protoporphyrin (ZnPP), a heme oxygenase-1 (HO-1) inhibitor, is potential to inhibit tumor proliferation and progression. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 48-52 26822586-5 2016 We hypothesize that ZnPP may modulate HIF-1alpha through inhibiting HO-1, and then inhibit angiogenesis and tumor progression. zinc protoporphyrin 20-24 heme oxygenase 1 Homo sapiens 68-72 26822586-11 2016 Moreover, we observed that the HO-1 inhibitor ZnPP inhibited the expressions of HIF-1alpha and VEGF coupled with cell proliferations of HCT-15 cells, suggesting that ZnPP blocked HIF-1alpha expression, and then inhibited the consequent VEGF production. zinc protoporphyrin 46-50 heme oxygenase 1 Homo sapiens 31-35 26822586-11 2016 Moreover, we observed that the HO-1 inhibitor ZnPP inhibited the expressions of HIF-1alpha and VEGF coupled with cell proliferations of HCT-15 cells, suggesting that ZnPP blocked HIF-1alpha expression, and then inhibited the consequent VEGF production. zinc protoporphyrin 166-170 heme oxygenase 1 Homo sapiens 31-35 26822586-14 2016 The HO-1 inhibitor, ZnPP, possessed the properties of anti-tumor agent by decreasing HIF-1alpha levels, blocking VEGF production, impairing tumor angiogenesis, and inhibiting tumor growth. zinc protoporphyrin 20-24 heme oxygenase 1 Homo sapiens 4-8 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 47-49 heme oxygenase 1 Homo sapiens 60-76 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 47-49 heme oxygenase 1 Homo sapiens 78-82 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 47-49 heme oxygenase 1 Homo sapiens 185-189 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. protoporphyrin IX 195-212 heme oxygenase 1 Homo sapiens 60-76 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. protoporphyrin IX 195-212 heme oxygenase 1 Homo sapiens 78-82 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. protoporphyrin IX 195-212 heme oxygenase 1 Homo sapiens 185-189 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 247-249 heme oxygenase 1 Homo sapiens 60-76 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 247-249 heme oxygenase 1 Homo sapiens 78-82 26655464-3 2016 In the present study, we demonstrate here that PB increased heme oxygenase-1 (HO-1) expression, which conferred protection against MPP(+)-induced cytotoxicity, because the inhibitor of HO-1 zinc protoporphyrin-IX attenuated the neuroprotection of PB. pinocembrin 247-249 heme oxygenase 1 Homo sapiens 185-189 26655464-6 2016 Taken together, the results suggest that PB enhances HO-1 expression to suppress MPP(+)-induced oxidative damage via ERK1/2 signaling pathways. pinocembrin 41-43 heme oxygenase 1 Homo sapiens 53-57 26655464-7 2016 These results revealed the mechanisms of PB enhances HO-1 expression, and contribute to shed some light on the mechanisms whereby PB inhibits the MPP(+)-induced neurotoxicity. pinocembrin 41-43 heme oxygenase 1 Homo sapiens 53-57 26834769-10 2015 Together, these results clearly suggest that HO-1 is involved in the alleviation of GA-induced PCD of drought-triggered rice aleurone layers by associating with NO. Gallium 84-86 heme oxygenase 1 Homo sapiens 45-49 26631587-9 2016 Key messages Bilirubin levels are inversely related to cardiovascular disease, and overexpression of heme oxygenase-1 (the enzyme that determines bilirubin production) has prevented post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and the progression of ventricular volumes and function in patients with acute myocardial infarction remained unexplored. Bilirubin 146-155 heme oxygenase 1 Homo sapiens 101-117 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Heme 79-83 heme oxygenase 1 Homo sapiens 18-22 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Biliverdine 87-105 heme oxygenase 1 Homo sapiens 18-22 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Carbon Monoxide 107-122 heme oxygenase 1 Homo sapiens 18-22 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Carbon Monoxide 124-126 heme oxygenase 1 Homo sapiens 18-22 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Iron 138-142 heme oxygenase 1 Homo sapiens 18-22 26834769-2 2015 Heme oxygenase-1 (HO-1) is known as a rate-liming enzyme in the degradation of heme to biliverdin IXalpha, carbon monoxide (CO), and free iron ions (Fe(2+)). Iron 149-151 heme oxygenase 1 Homo sapiens 18-22 26834769-4 2015 Our previous studies confirmed that HO-1 inducer hematin (Ht) promotes the germination of rice seeds in drought (20% polyethylene glycol-6000, PEG) conditions, but the corresponding effects of HO-1 on the alleviation of germination-triggered PCD in GA-treated rice aleurone layers remain unknown. Polyethylene Glycol 6000 117-141 heme oxygenase 1 Homo sapiens 36-40 26834769-4 2015 Our previous studies confirmed that HO-1 inducer hematin (Ht) promotes the germination of rice seeds in drought (20% polyethylene glycol-6000, PEG) conditions, but the corresponding effects of HO-1 on the alleviation of germination-triggered PCD in GA-treated rice aleurone layers remain unknown. Polyethylene Glycol 6000 143-146 heme oxygenase 1 Homo sapiens 36-40 26834769-5 2015 The present study has determined that GA co-treated with PEG results in lower HO-1 transcript levels and HO activity, which in turn results in the development of vacuoles in aleurone cells, followed by PCD. Gallium 38-40 heme oxygenase 1 Homo sapiens 78-82 26834769-5 2015 The present study has determined that GA co-treated with PEG results in lower HO-1 transcript levels and HO activity, which in turn results in the development of vacuoles in aleurone cells, followed by PCD. Polyethylene Glycol 6000 57-60 heme oxygenase 1 Homo sapiens 78-82 26834769-6 2015 The pharmacology approach illustrated that up- or down-regulated HO-1 gene expression and HO activity delayed or accelerated GA-induced PCD. Gallium 125-127 heme oxygenase 1 Homo sapiens 65-69 26834769-7 2015 Furthermore, the application of the HO-1 inducer Ht and nitric oxide (NO) donor sodium nitroprusside (SNP) not only activated HO-1 gene expression, HO activity, and endogenous NO content, but also blocked GA-induced rapid vacuolation and accelerated aleurone layers PCD under drought stress. Nitric Oxide 56-68 heme oxygenase 1 Homo sapiens 36-40 26834769-7 2015 Furthermore, the application of the HO-1 inducer Ht and nitric oxide (NO) donor sodium nitroprusside (SNP) not only activated HO-1 gene expression, HO activity, and endogenous NO content, but also blocked GA-induced rapid vacuolation and accelerated aleurone layers PCD under drought stress. Nitric Oxide 56-68 heme oxygenase 1 Homo sapiens 126-130 26834769-7 2015 Furthermore, the application of the HO-1 inducer Ht and nitric oxide (NO) donor sodium nitroprusside (SNP) not only activated HO-1 gene expression, HO activity, and endogenous NO content, but also blocked GA-induced rapid vacuolation and accelerated aleurone layers PCD under drought stress. Nitroprusside 80-100 heme oxygenase 1 Homo sapiens 36-40 26834769-7 2015 Furthermore, the application of the HO-1 inducer Ht and nitric oxide (NO) donor sodium nitroprusside (SNP) not only activated HO-1 gene expression, HO activity, and endogenous NO content, but also blocked GA-induced rapid vacuolation and accelerated aleurone layers PCD under drought stress. Nitroprusside 80-100 heme oxygenase 1 Homo sapiens 126-130 26834769-8 2015 However, both HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) and NO scavenger 2-(4-carboxyphenyl0-4, 4,5,5-tetramethylimidazoline-l-oxyl-3-oxide potassium salt (cPTIO) reserved the effects of Ht and SNP on rice aleurone layer PCD under drought stress by down-regulating endogenous HO-1 and NO, respectively. zinc protoporphyrin 29-51 heme oxygenase 1 Homo sapiens 14-18 26834769-8 2015 However, both HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) and NO scavenger 2-(4-carboxyphenyl0-4, 4,5,5-tetramethylimidazoline-l-oxyl-3-oxide potassium salt (cPTIO) reserved the effects of Ht and SNP on rice aleurone layer PCD under drought stress by down-regulating endogenous HO-1 and NO, respectively. zinc protoporphyrin 53-59 heme oxygenase 1 Homo sapiens 14-18 26834769-8 2015 However, both HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) and NO scavenger 2-(4-carboxyphenyl0-4, 4,5,5-tetramethylimidazoline-l-oxyl-3-oxide potassium salt (cPTIO) reserved the effects of Ht and SNP on rice aleurone layer PCD under drought stress by down-regulating endogenous HO-1 and NO, respectively. 2-(4-carboxyphenyl0-4, 4,5,5-tetramethylimidazoline-l-oxyl-3-oxide potassium salt 78-159 heme oxygenase 1 Homo sapiens 281-285 26652036-6 2016 Furthermore, heme hydroxylation proceeds three times more slowly, and the oxy-Fe(II) state is 100-fold less stable in HO2 than in HO1. oxy-fe(ii) 74-84 heme oxygenase 1 Homo sapiens 130-133 26652036-0 2016 Comparison of the Mechanisms of Heme Hydroxylation by Heme Oxygenases-1 and -2: Kinetic and Cryoreduction Studies. Heme 32-36 heme oxygenase 1 Homo sapiens 54-78 26652036-1 2016 The two isoforms of human heme oxygenase (HO1 and HO2) catalyze oxidative degradation of heme to biliverdin, Fe, and CO. Heme 26-30 heme oxygenase 1 Homo sapiens 42-45 26652036-1 2016 The two isoforms of human heme oxygenase (HO1 and HO2) catalyze oxidative degradation of heme to biliverdin, Fe, and CO. Biliverdine 97-107 heme oxygenase 1 Homo sapiens 42-45 26652036-1 2016 The two isoforms of human heme oxygenase (HO1 and HO2) catalyze oxidative degradation of heme to biliverdin, Fe, and CO. Iron 109-111 heme oxygenase 1 Homo sapiens 42-45 26652036-1 2016 The two isoforms of human heme oxygenase (HO1 and HO2) catalyze oxidative degradation of heme to biliverdin, Fe, and CO. Carbon Monoxide 117-119 heme oxygenase 1 Homo sapiens 42-45 26779023-0 2015 Ferulic Acid Regulates the Nrf2/Heme Oxygenase-1 System and Counteracts Trimethyltin-Induced Neuronal Damage in the Human Neuroblastoma Cell Line SH-SY5Y. ferulic acid 0-12 heme oxygenase 1 Homo sapiens 32-48 26779023-1 2015 Over the past years, several lines of evidence have pointed out the efficacy of ferulic acid (FA) in counteracting oxidative stress elicited by beta-amyloid or free radical initiators, based on the ability of this natural antioxidant to up-regulate the heme oxygenase-1 (HO-1) and biliverdin reductase (BVR) system. ferulic acid 80-92 heme oxygenase 1 Homo sapiens 253-269 26779023-1 2015 Over the past years, several lines of evidence have pointed out the efficacy of ferulic acid (FA) in counteracting oxidative stress elicited by beta-amyloid or free radical initiators, based on the ability of this natural antioxidant to up-regulate the heme oxygenase-1 (HO-1) and biliverdin reductase (BVR) system. ferulic acid 80-92 heme oxygenase 1 Homo sapiens 271-275 26779023-9 2015 All together, these results corroborate the neuroprotective effect of FA through the up-regulation of the HO-1/BVR system, via carbon monoxide and BR formation, and provide the first evidence on the role of HO-1/Nrf2 axis in FA-related enhancement of cell stress response in human neurons. Carbon Monoxide 127-142 heme oxygenase 1 Homo sapiens 106-114 26779023-9 2015 All together, these results corroborate the neuroprotective effect of FA through the up-regulation of the HO-1/BVR system, via carbon monoxide and BR formation, and provide the first evidence on the role of HO-1/Nrf2 axis in FA-related enhancement of cell stress response in human neurons. Carbon Monoxide 127-142 heme oxygenase 1 Homo sapiens 106-110 26779023-9 2015 All together, these results corroborate the neuroprotective effect of FA through the up-regulation of the HO-1/BVR system, via carbon monoxide and BR formation, and provide the first evidence on the role of HO-1/Nrf2 axis in FA-related enhancement of cell stress response in human neurons. Bilirubin 147-149 heme oxygenase 1 Homo sapiens 106-114 26779023-9 2015 All together, these results corroborate the neuroprotective effect of FA through the up-regulation of the HO-1/BVR system, via carbon monoxide and BR formation, and provide the first evidence on the role of HO-1/Nrf2 axis in FA-related enhancement of cell stress response in human neurons. Bilirubin 147-149 heme oxygenase 1 Homo sapiens 106-110 26430779-7 2016 The sTNF-alphaR-Fc/HO-1 group showed the best glucose tolerance. Glucose 46-53 heme oxygenase 1 Homo sapiens 19-23 26631587-9 2016 Key messages Bilirubin levels are inversely related to cardiovascular disease, and overexpression of heme oxygenase-1 (the enzyme that determines bilirubin production) has prevented post-infarction ventricular remodeling in experimental animals, but the association between bilirubin levels and the progression of ventricular volumes and function in patients with acute myocardial infarction remained unexplored. Bilirubin 274-283 heme oxygenase 1 Homo sapiens 101-117 26759705-4 2016 Furthermore, RA increased the expression and activity of superoxide dismutase, catalase, heme oxygenase-1, and their transcription factor Nrf2, which are decreased by UVB radiation. rosmarinic acid 13-15 heme oxygenase 1 Homo sapiens 89-105 27251503-3 2016 Although hemin treatment caused the increased expression of heme oxygenase (HO)-1, the inhibition of HO activity resulted in the aggravation of hemin-induced barrier dysfunction. Hemin 9-14 heme oxygenase 1 Homo sapiens 60-81 27080946-0 2016 Tanshinone IIA Induces Heme Oxygenase 1 Expression and Inhibits Cyclic Strain-Induced Interleukin 8 Expression in Vascular Endothelial Cells. tanshinone 0-10 heme oxygenase 1 Homo sapiens 23-39 27080946-6 2016 HO-1 silencing significantly abrogated the repressive effects of tanshinone IIA on strain-induced IL-8 expression, which suggests HO-1 has a role in mediating the effects of tanshinone IIA. tanshinone 65-75 heme oxygenase 1 Homo sapiens 0-4 27080946-6 2016 HO-1 silencing significantly abrogated the repressive effects of tanshinone IIA on strain-induced IL-8 expression, which suggests HO-1 has a role in mediating the effects of tanshinone IIA. tanshinone 65-75 heme oxygenase 1 Homo sapiens 130-134 27080946-6 2016 HO-1 silencing significantly abrogated the repressive effects of tanshinone IIA on strain-induced IL-8 expression, which suggests HO-1 has a role in mediating the effects of tanshinone IIA. tanshinone 65-79 heme oxygenase 1 Homo sapiens 0-4 27080946-6 2016 HO-1 silencing significantly abrogated the repressive effects of tanshinone IIA on strain-induced IL-8 expression, which suggests HO-1 has a role in mediating the effects of tanshinone IIA. tanshinone 65-79 heme oxygenase 1 Homo sapiens 130-134 27080946-7 2016 This study reports for the first time that tanshinone IIA inhibits cyclic strain-induced IL-8 expression via the induction of HO-1 in endothelial cells, providing valuable new insight into the molecular pathways that may contribute to the effects of tanshinone IIA. tanshinone 43-53 heme oxygenase 1 Homo sapiens 126-130 27080946-7 2016 This study reports for the first time that tanshinone IIA inhibits cyclic strain-induced IL-8 expression via the induction of HO-1 in endothelial cells, providing valuable new insight into the molecular pathways that may contribute to the effects of tanshinone IIA. tanshinone 250-260 heme oxygenase 1 Homo sapiens 126-130 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. Technetium 29-31 heme oxygenase 1 Homo sapiens 97-118 26277982-5 2016 In addition, DMHC induced HO-1 expression as well as NRF2 activation. dmhc 13-17 heme oxygenase 1 Homo sapiens 26-30 26277982-6 2016 Furthermore, HO-1 knockdown using siRNA reversed the inhibitory effect of DMHC on TNF-alpha-induced ICAM-1 expression and adhesion of monocytes to keratinocytes. dmhc 74-78 heme oxygenase 1 Homo sapiens 13-17 26277982-7 2016 These results suggest that DMHC may inhibit TNF-alpha-induced ICAM-1 expression and adhesion of monocytes to keratinocytes by suppressing the signaling cascades leading to NF-kappaB activation and inducing HO-1 expression in keratinocytes. dmhc 27-31 heme oxygenase 1 Homo sapiens 206-210 27412411-1 2016 Heme oxygenase 1 (HO-1) is an inducible stress-response enzyme that not only catalyzes the degradation of heme (e.g., released from erythrocytes) but also has an important function in various physiological and pathophysiological states associated with cellular stress, such as ischemic/reperfusion injury. Heme 106-110 heme oxygenase 1 Homo sapiens 0-16 27412411-1 2016 Heme oxygenase 1 (HO-1) is an inducible stress-response enzyme that not only catalyzes the degradation of heme (e.g., released from erythrocytes) but also has an important function in various physiological and pathophysiological states associated with cellular stress, such as ischemic/reperfusion injury. Heme 106-110 heme oxygenase 1 Homo sapiens 18-22 26277982-0 2016 2,3-Dimethoxy-2"-hydroxychalcone ameliorates TNF-alpha-induced ICAM-1 expression and subsequent monocyte adhesiveness via NF-kappaB inhibition and HO-1 induction in HaCaT cells. 2,3-dimethoxy-2'-hydroxychalcone 0-32 heme oxygenase 1 Homo sapiens 147-151 27322610-6 2016 HO-1 knockdown experiments were done to confirm its role in the action of periostin in high glucose-exposed HUVECs. Glucose 92-99 heme oxygenase 1 Homo sapiens 0-4 27322610-11 2016 HO-1 silencing restored high glucose-induced ROS generation and apoptotic response in periostin-overexpressing HUVECs. Glucose 29-36 heme oxygenase 1 Homo sapiens 0-4 27322610-11 2016 HO-1 silencing restored high glucose-induced ROS generation and apoptotic response in periostin-overexpressing HUVECs. Reactive Oxygen Species 45-48 heme oxygenase 1 Homo sapiens 0-4 27322610-12 2016 CONCLUSION: Periostin mitigates high glucose-induced mitochondrial apoptosis in endothelial cells, via activation of Nrf2/HO-1 signaling and reduction of ROS formation. Glucose 37-44 heme oxygenase 1 Homo sapiens 122-126 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. Technetium 29-31 heme oxygenase 1 Homo sapiens 136-140 27412411-7 2016 Our results indicate that HO-1 is an inhibitor of hematopoietic cell migration in response to crucial BM homing chemoattractants such as stromal-derived factor 1 (SDF-1) and sphingosine-1-phosphate (S1P). sphingosine 1-phosphate 174-197 heme oxygenase 1 Homo sapiens 26-30 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. zinc protoporphyrin 50-72 heme oxygenase 1 Homo sapiens 97-118 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. zinc protoporphyrin 50-72 heme oxygenase 1 Homo sapiens 136-140 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. zinc protoporphyrin 74-78 heme oxygenase 1 Homo sapiens 97-118 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. zinc protoporphyrin 74-78 heme oxygenase 1 Homo sapiens 136-140 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. Technetium 186-188 heme oxygenase 1 Homo sapiens 97-118 27383847-6 2016 All those effects induced by TC were inhibited by zinc protoporphyrin IX (ZnPP), an inhibitor of heme oxygenase (HO)-1, indicating that HO-1 was responsible the anti-psoriatic effect of TC. Technetium 186-188 heme oxygenase 1 Homo sapiens 136-140 27383847-9 2016 Overexpression of p65 NF-x03BA;B significantly suppressed TC-induced decrease of TNFalpha, IL-17A and IL-23 expression and keratinocyte proliferation, indicating that HO-1-mediated downregulation of NF-x03BA;B was involved in the anti-psoriatic effect of TC. Technetium 58-60 heme oxygenase 1 Homo sapiens 167-171 27383847-9 2016 Overexpression of p65 NF-x03BA;B significantly suppressed TC-induced decrease of TNFalpha, IL-17A and IL-23 expression and keratinocyte proliferation, indicating that HO-1-mediated downregulation of NF-x03BA;B was involved in the anti-psoriatic effect of TC. Technetium 255-257 heme oxygenase 1 Homo sapiens 167-171 26865999-5 2016 Cultures of normal human melanocytes were treated with the HO-1 inducer cobalt protoporphyrin (CoPP) or the HO-1 inhibitor zinc protoporphyrin (ZnPP). zinc protoporphyrin 123-142 heme oxygenase 1 Homo sapiens 108-112 26573721-0 2016 Vanadium(III)-L-cysteine protects cisplatin-induced nephropathy through activation of Nrf2/HO-1 pathway. vanadium(III)-cysteine 0-24 heme oxygenase 1 Homo sapiens 91-95 27558512-5 2016 Macrophage treatment with the iron oxide NPs showed a dose-dependent increase of heme oxygenase 1 (HO-1) and NAD(P)H:quinone oxidoreductase (NQO-1). ferric oxide 30-40 heme oxygenase 1 Homo sapiens 81-97 26573721-0 2016 Vanadium(III)-L-cysteine protects cisplatin-induced nephropathy through activation of Nrf2/HO-1 pathway. Cisplatin 34-43 heme oxygenase 1 Homo sapiens 91-95 26573721-11 2016 VC-III administration also stimulated Nrf2-mediated antioxidant defense system by promotion of downstream antioxidant enzymes, such as HO-1. vc-iii 0-6 heme oxygenase 1 Homo sapiens 135-139 27764834-10 2016 Nuclear Nrf2 protein and HO-1 mRNA levels were both increased in the HTR-8/SVneo cell line following stimulation with tBHQ (p < 0.05). 2-tert-butylhydroquinone 118-122 heme oxygenase 1 Homo sapiens 25-29 26602157-4 2016 Among the genes upregulated in response to manumycin were HMOX-1, TNFRSF10A, IL-1R1, TICAM2, NLRP12 after 4h and only IL-1R1 after 8h. manumycin 43-52 heme oxygenase 1 Homo sapiens 58-64 26386740-0 2016 Chlorogenic acid improves ex vivo vessel function and protects endothelial cells against HOCl-induced oxidative damage, via increased production of nitric oxide and induction of Hmox-1. Chlorogenic Acid 0-16 heme oxygenase 1 Homo sapiens 178-184 26403645-6 2016 Knockdown of p62, quinone oxidoreductase-1, heme oxygenase-1, and ferritin heavy chain-1 by RNA interference in HCC cells promoted ferroptosis in response to erastin and sorafenib. erastin 158-165 heme oxygenase 1 Homo sapiens 18-60 26403645-6 2016 Knockdown of p62, quinone oxidoreductase-1, heme oxygenase-1, and ferritin heavy chain-1 by RNA interference in HCC cells promoted ferroptosis in response to erastin and sorafenib. Sorafenib 170-179 heme oxygenase 1 Homo sapiens 18-60 26302870-1 2016 Previous studies indicated the potential of zinc protoporphyrin (ZnPP) as an antitumor agent targeting to the tumor survival factor heme oxygenase-1, and/or for photodynamic therapy (PDT). zinc protoporphyrin 44-63 heme oxygenase 1 Homo sapiens 132-148 26302870-1 2016 Previous studies indicated the potential of zinc protoporphyrin (ZnPP) as an antitumor agent targeting to the tumor survival factor heme oxygenase-1, and/or for photodynamic therapy (PDT). zinc protoporphyrin 65-69 heme oxygenase 1 Homo sapiens 132-148 26058377-9 2016 D006 promoted the expression of HO-1 protein in a time-dependent manner while the HO-1 inhibitor, Znpp, reversed the protective effects of D006. d006 0-4 heme oxygenase 1 Homo sapiens 32-36 26058377-9 2016 D006 promoted the expression of HO-1 protein in a time-dependent manner while the HO-1 inhibitor, Znpp, reversed the protective effects of D006. zinc protoporphyrin 98-102 heme oxygenase 1 Homo sapiens 82-86 26058377-9 2016 D006 promoted the expression of HO-1 protein in a time-dependent manner while the HO-1 inhibitor, Znpp, reversed the protective effects of D006. d006 139-143 heme oxygenase 1 Homo sapiens 82-86 26058377-11 2016 In conclusion, our results indicate that a new DSS derivative exhibits promising protective effects against Dox-induced cardiotoxicity both in vivo and in vitro, an effect at least partially mediated by induction of HO-1 expression and the activation of mitochondrial biogenesis. 3,4-dihydroxyphenyllactic acid 47-50 heme oxygenase 1 Homo sapiens 216-220 26058377-11 2016 In conclusion, our results indicate that a new DSS derivative exhibits promising protective effects against Dox-induced cardiotoxicity both in vivo and in vitro, an effect at least partially mediated by induction of HO-1 expression and the activation of mitochondrial biogenesis. Doxorubicin 108-111 heme oxygenase 1 Homo sapiens 216-220 26630500-5 2016 Additionally, these Pb concentrations induced oxidative stress in HepG2 cells in terms of production of reactive oxygen species (ROS) and induced heme oxygenase-1 mRNA expression in a concentration-dependent phenomenon. Lead 20-22 heme oxygenase 1 Homo sapiens 146-162 26386740-0 2016 Chlorogenic acid improves ex vivo vessel function and protects endothelial cells against HOCl-induced oxidative damage, via increased production of nitric oxide and induction of Hmox-1. Hypochlorous Acid 89-93 heme oxygenase 1 Homo sapiens 178-184 27110594-4 2016 Relative to WT mice, mice globally overexpressing human HO-1 were protected from DOX-induced dilated cardiomyopathy, cardiac cytoarchitectural derangement, and infiltration of CD11b+ mononuclear phagocytes. Doxorubicin 81-84 heme oxygenase 1 Homo sapiens 56-60 27057275-0 2016 Synergistic Effects of Cilostazol and Probucol on ER Stress-Induced Hepatic Steatosis via Heme Oxygenase-1-Dependent Activation of Mitochondrial Biogenesis. Cilostazol 23-33 heme oxygenase 1 Homo sapiens 90-106 26542248-6 2016 We further demonstrated that HO-1 could be induced by PbAc through the P38, ERK1/2, and PI3K/AKT signaling pathways in a ROS-dependent manner and through the JNK pathway in a ROS-independent manner. Reactive Oxygen Species 121-124 heme oxygenase 1 Homo sapiens 29-33 26542248-6 2016 We further demonstrated that HO-1 could be induced by PbAc through the P38, ERK1/2, and PI3K/AKT signaling pathways in a ROS-dependent manner and through the JNK pathway in a ROS-independent manner. Reactive Oxygen Species 175-178 heme oxygenase 1 Homo sapiens 29-33 26542248-7 2016 Further investigation revealed that HO-1 overexpression significantly restrained cell apoptosis and ROS production induced by PbAc in SH-SY5Y cells. Reactive Oxygen Species 100-103 heme oxygenase 1 Homo sapiens 36-40 26542248-8 2016 Moreover, HO-1 knockdown aggravated PbAc-induced cell apoptosis and ROS production. Reactive Oxygen Species 68-71 heme oxygenase 1 Homo sapiens 10-14 26798413-0 2016 t-BHQ Provides Protection against Lead Neurotoxicity via Nrf2/HO-1 Pathway. 2-tert-butylhydroquinone 0-5 heme oxygenase 1 Homo sapiens 62-66 26798413-10 2016 However, knockdown of Nrf2 or HO-1 adversely affected the protective effects of t-BHQ against lead toxicity in SH-SY5Y cells. 2-tert-butylhydroquinone 80-85 heme oxygenase 1 Homo sapiens 30-34 26798413-11 2016 Thus, t-BHQ can protect against lead neurotoxicity, depending on the Nrf2/HO-1 pathway. 2-tert-butylhydroquinone 6-11 heme oxygenase 1 Homo sapiens 74-78 27057275-0 2016 Synergistic Effects of Cilostazol and Probucol on ER Stress-Induced Hepatic Steatosis via Heme Oxygenase-1-Dependent Activation of Mitochondrial Biogenesis. Probucol 38-46 heme oxygenase 1 Homo sapiens 90-106 26765462-4 2016 Addition of ALA strongly attenuated ROS production and further increased HO-1 expression. Thioctic Acid 12-15 heme oxygenase 1 Homo sapiens 73-77 26765462-5 2016 However, by pretreatment of zinc protoporphyrin (ZnPP), HO-1 inhibitor, ALA inhibition of ROS generation by H2O2 was abolished. Thioctic Acid 72-75 heme oxygenase 1 Homo sapiens 56-60 26765462-5 2016 However, by pretreatment of zinc protoporphyrin (ZnPP), HO-1 inhibitor, ALA inhibition of ROS generation by H2O2 was abolished. Reactive Oxygen Species 90-93 heme oxygenase 1 Homo sapiens 56-60 26765462-5 2016 However, by pretreatment of zinc protoporphyrin (ZnPP), HO-1 inhibitor, ALA inhibition of ROS generation by H2O2 was abolished. Hydrogen Peroxide 108-112 heme oxygenase 1 Homo sapiens 56-60 27034731-3 2016 The results showed that these anthocyanins decreased the levels of ROS and XO-1 but increased the levels of SOD and HO-1. Anthocyanins 30-42 heme oxygenase 1 Homo sapiens 116-120 27313828-7 2016 SPRC-induced HO-1 expression and Nrf2 translocation were abolished by the phosphoinositide 3-kinase (PI3K) inhibitor LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 117-125 heme oxygenase 1 Homo sapiens 13-17 26680374-1 2016 BACKGROUND: The enzyme heme oxygenase-1 (HO-1) degrades heme and protects against ischemia-reperfusion injury. Heme 23-27 heme oxygenase 1 Homo sapiens 41-45 27313828-8 2016 Moreover, the antioxidative effect of SPRC was abolished by CSE inhibitor DL-propargylglycine (PAG) and HO-1 siRNA. S-propargylcysteine 38-42 heme oxygenase 1 Homo sapiens 104-108 27313828-9 2016 Therefore, these results proved that SPRC produced an antioxidative effect on NAFLD through the PI3K/Akt/Nrf2/HO-1 signaling pathway. S-propargylcysteine 37-41 heme oxygenase 1 Homo sapiens 110-114 27050899-7 2016 We demonstrated that nicorandil increased HO-1 expression in HUVECs. Nicorandil 21-31 heme oxygenase 1 Homo sapiens 42-46 27050899-8 2016 In addition, cobalt protoporphyrin (10 mumol/l), an inducer of HO-1 expression, mimicked the effects of nicorandil and inhibited IL-8 expression under cyclic strain, whereas zinc protoporphyrin IX (10 mumol/l), an inhibitor of HO-1 expression, antagonized the effect of nicorandil. cobaltiprotoporphyrin 13-34 heme oxygenase 1 Homo sapiens 63-67 27050899-8 2016 In addition, cobalt protoporphyrin (10 mumol/l), an inducer of HO-1 expression, mimicked the effects of nicorandil and inhibited IL-8 expression under cyclic strain, whereas zinc protoporphyrin IX (10 mumol/l), an inhibitor of HO-1 expression, antagonized the effect of nicorandil. cobaltiprotoporphyrin 13-34 heme oxygenase 1 Homo sapiens 227-231 27050899-8 2016 In addition, cobalt protoporphyrin (10 mumol/l), an inducer of HO-1 expression, mimicked the effects of nicorandil and inhibited IL-8 expression under cyclic strain, whereas zinc protoporphyrin IX (10 mumol/l), an inhibitor of HO-1 expression, antagonized the effect of nicorandil. Nicorandil 104-114 heme oxygenase 1 Homo sapiens 63-67 27050899-9 2016 HO-1 silencing significantly abrogated the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression, suggesting that HO-1 plays a role in the mechanism of action of nicorandil. Nicorandil 65-75 heme oxygenase 1 Homo sapiens 0-4 27050899-9 2016 HO-1 silencing significantly abrogated the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression, suggesting that HO-1 plays a role in the mechanism of action of nicorandil. Nicorandil 65-75 heme oxygenase 1 Homo sapiens 134-138 27050899-9 2016 HO-1 silencing significantly abrogated the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression, suggesting that HO-1 plays a role in the mechanism of action of nicorandil. Nicorandil 182-192 heme oxygenase 1 Homo sapiens 0-4 27050899-9 2016 HO-1 silencing significantly abrogated the inhibitory effects of nicorandil on cyclic strain-induced IL-8 expression, suggesting that HO-1 plays a role in the mechanism of action of nicorandil. Nicorandil 182-192 heme oxygenase 1 Homo sapiens 134-138 27050899-10 2016 KEY MESSAGES: This study is the first to report that nicorandil inhibits cyclic strain-induced IL-8 expression through the induction of HO-1 expression in HUVECs. Nicorandil 53-63 heme oxygenase 1 Homo sapiens 136-140 26681956-2 2016 To examine the mechanisms by which this occurs, we hypothesized that an increase in heme oxygenase 1, a potent antioxidant gene, will decrease uric acid levels and adipocyte dysfunction via suppression of ROS and xanthine oxidase (XO) levels. Uric Acid 143-152 heme oxygenase 1 Homo sapiens 84-100 26681956-2 2016 To examine the mechanisms by which this occurs, we hypothesized that an increase in heme oxygenase 1, a potent antioxidant gene, will decrease uric acid levels and adipocyte dysfunction via suppression of ROS and xanthine oxidase (XO) levels. ros 205-208 heme oxygenase 1 Homo sapiens 84-100 26515032-4 2016 Recent reports indicate that HO-1/HO-2 protein expression and HO activity have several important roles in hemostasis and reactive oxygen species (ROS)-dependent perturbations associated with metabolic syndrome. Reactive Oxygen Species 121-144 heme oxygenase 1 Homo sapiens 29-38 26515032-4 2016 Recent reports indicate that HO-1/HO-2 protein expression and HO activity have several important roles in hemostasis and reactive oxygen species (ROS)-dependent perturbations associated with metabolic syndrome. Reactive Oxygen Species 146-149 heme oxygenase 1 Homo sapiens 29-38 27313828-0 2016 S-Propargyl-cysteine Exerts a Novel Protective Effect on Methionine and Choline Deficient Diet-Induced Fatty Liver via Akt/Nrf2/HO-1 Pathway. S-propargylcysteine 0-20 heme oxygenase 1 Homo sapiens 128-132 27578289-3 2016 In the present study, we have investigated the effects of bortezomib, a clinically used proteasome inhibitor, on the regulation of HO-1 and COX-2 in cultured human microvascular endothelial cells (HMECs). Bortezomib 58-68 heme oxygenase 1 Homo sapiens 131-135 27578289-4 2016 Bortezomib treatment of HMECS induced dose- and time-dependent expression of HO-1 and COX-2 mRNA and protein, and triggered nuclear translocation of nuclear factor erythroid 2-related transcription factor (Nrf2). Bortezomib 0-10 heme oxygenase 1 Homo sapiens 77-81 27578289-5 2016 These findings suggest that HO-1/COX-2-mediated induction of antioxidant mechanisms via Nrf2 activation may contribute to the cytoprotective effects of bortezomib in microvascular endothelium. Bortezomib 152-162 heme oxygenase 1 Homo sapiens 28-32 28124024-0 2016 Iron Supplementation Alters Heme and Heme Oxygenase 1 (HO-1) Levels In Pregnant Women in Ghana. Iron 0-4 heme oxygenase 1 Homo sapiens 37-53 28124024-0 2016 Iron Supplementation Alters Heme and Heme Oxygenase 1 (HO-1) Levels In Pregnant Women in Ghana. Iron 0-4 heme oxygenase 1 Homo sapiens 55-59 28124024-4 2016 We hypothesized that pregnant women with malaria who took iron supplements will have higher levels of Heme/HO-1 than those who did not take iron supplements. Iron 58-62 heme oxygenase 1 Homo sapiens 107-111 26166253-2 2016 HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXalpha (BV), the latter which is converted to bilirubin-IXalpha (BR). Carbon Monoxide 132-147 heme oxygenase 1 Homo sapiens 0-4 26166253-2 2016 HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXalpha (BV), the latter which is converted to bilirubin-IXalpha (BR). Carbon Monoxide 149-151 heme oxygenase 1 Homo sapiens 0-4 26166253-2 2016 HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXalpha (BV), the latter which is converted to bilirubin-IXalpha (BR). Iron 154-158 heme oxygenase 1 Homo sapiens 0-4 26166253-2 2016 HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXalpha (BV), the latter which is converted to bilirubin-IXalpha (BR). Biliverdine 164-182 heme oxygenase 1 Homo sapiens 0-4 26166253-2 2016 HO-1, a cellular stress protein, serves a vital metabolic function as the rate-limiting step in the degradation of heme to generate carbon monoxide (CO), iron, and biliverdin-IXalpha (BV), the latter which is converted to bilirubin-IXalpha (BR). Bilirubin 222-239 heme oxygenase 1 Homo sapiens 0-4 29668174-0 2016 Induction of Heme oxygenase 1 protects hepatocytes from Isoniazid - Rifampicin induced cell death: an in vitro study. isoniazid - rifampicin 56-78 heme oxygenase 1 Homo sapiens 13-29 29668174-5 2016 The E47 cells were treated with hemin chloride to induce HO-1 expression during INH-RMP treatment. Hemin 32-46 heme oxygenase 1 Homo sapiens 57-61 29668174-10 2016 Addition of Hemin chloride during treatment of INH-RMP induced HO-1 in E47 cells and reversed the drug induced liver injury. Hemin 12-26 heme oxygenase 1 Homo sapiens 63-67 29668174-11 2016 Silencing the HO-1 gene using siRNA potentiated INH-RMP induced cell death of the E47 cells Conclusion: Induction of HO-1 ameliorated INH-RMP induced cell death of hepatocytes. inh-rmp 48-55 heme oxygenase 1 Homo sapiens 14-18 29668174-11 2016 Silencing the HO-1 gene using siRNA potentiated INH-RMP induced cell death of the E47 cells Conclusion: Induction of HO-1 ameliorated INH-RMP induced cell death of hepatocytes. inh-rmp 48-55 heme oxygenase 1 Homo sapiens 117-121 26680374-3 2016 Heme arginate (HA) safely induces HO-1 in humans. heme arginate 0-13 heme oxygenase 1 Homo sapiens 34-38 26680374-3 2016 Heme arginate (HA) safely induces HO-1 in humans. heme arginate 15-17 heme oxygenase 1 Homo sapiens 34-38 26680374-12 2016 Heme arginate increased day 5 tissue HO-1 protein immunopositivity compared with placebo: HA 0.21 versus placebo -0.03 (P = 0.02) and % HO-1-positive renal macrophage also increased: HA 50.8 cells per high power field versus placebo 22.3 (P = 0.012). heme arginate 0-13 heme oxygenase 1 Homo sapiens 37-41 26680374-12 2016 Heme arginate increased day 5 tissue HO-1 protein immunopositivity compared with placebo: HA 0.21 versus placebo -0.03 (P = 0.02) and % HO-1-positive renal macrophage also increased: HA 50.8 cells per high power field versus placebo 22.3 (P = 0.012). heme arginate 0-13 heme oxygenase 1 Homo sapiens 136-140 27725392-5 2016 Under the gaseous phase administration, acetic acid and mixed acids caused a slight increase, decrease and increase on the interleukin-8 production, the mRNA expression of the heme oxygenase-1 (HO-1) gene and the HO-1 production, respectively, at one or more time points. Acetic Acid 40-51 heme oxygenase 1 Homo sapiens 176-192 27725392-5 2016 Under the gaseous phase administration, acetic acid and mixed acids caused a slight increase, decrease and increase on the interleukin-8 production, the mRNA expression of the heme oxygenase-1 (HO-1) gene and the HO-1 production, respectively, at one or more time points. Acetic Acid 40-51 heme oxygenase 1 Homo sapiens 194-198 27725392-5 2016 Under the gaseous phase administration, acetic acid and mixed acids caused a slight increase, decrease and increase on the interleukin-8 production, the mRNA expression of the heme oxygenase-1 (HO-1) gene and the HO-1 production, respectively, at one or more time points. Acetic Acid 40-51 heme oxygenase 1 Homo sapiens 213-217 26670903-11 2015 CONCLUSION: In summary, the present study showed that HO-1 attenuates the Cd-induced caspase 3 dependent pathway of apoptosis in HepG2 cells, probably by modulating Cd-induced oxidative stress. Cadmium 74-76 heme oxygenase 1 Homo sapiens 54-58 26690352-6 2015 Pharmacological induction of HO-1 with cobalt-protoporphyrin IX reduced, whereas inhibition of HO-1 with either zinc-protoporphyrin IX or siRNA-mediated knockdown increased HLA I Ab-dependent up-regulation of VCAM-1. cobaltiprotoporphyrin 39-63 heme oxygenase 1 Homo sapiens 29-33 26690352-6 2015 Pharmacological induction of HO-1 with cobalt-protoporphyrin IX reduced, whereas inhibition of HO-1 with either zinc-protoporphyrin IX or siRNA-mediated knockdown increased HLA I Ab-dependent up-regulation of VCAM-1. zinc protoporphyrin 112-134 heme oxygenase 1 Homo sapiens 95-99 26782424-0 2015 Role of heme oxygenase-1 in demethylating effects on SKM-1 cells induced by decitabine. Decitabine 76-86 heme oxygenase 1 Homo sapiens 8-24 26782424-1 2015 We evaluated the influence of heme oxygenase-1 (HO-1) gene inhibition in myelodysplastic syndrome (MDS) cell line SKM-1 on enhancement of the demethylating effects of decitabine on p15, and explored the possible mechanism. Decitabine 167-177 heme oxygenase 1 Homo sapiens 48-52 26782424-7 2015 CCK-8 assay showed that after HO-1 gene expression was inhibited; the proliferation rate of SKM-1 cells treated by decitabine (70.91 +- 0.05%) was significantly higher than that of the control group (53.67 +- 0.05%). Decitabine 115-125 heme oxygenase 1 Homo sapiens 30-34 26782424-8 2015 Flow cytometry showed that the apoptotic rate of SKM- 1 cells treated by decitabine in combination with HO-1 expression inhibition (44.25 +- 0.05%) exceeded that of the cells treated by this drug alone (37.70 +- 0.05%). Decitabine 73-83 heme oxygenase 1 Homo sapiens 104-108 26782424-10 2015 As suggested by western blot, the degree of methylation of the p15 protein was changed after decitabine treatment when the expression of the HO-1 protein was changed, being associated with the affected DNMT1 expression. Decitabine 93-103 heme oxygenase 1 Homo sapiens 141-145 26782424-11 2015 Inhibited HO-1 expression attenuated the hypermethylation of CDKN2B by suppressing DNMT1, which was conducive to treatment by cooperating with decitabine. Decitabine 143-153 heme oxygenase 1 Homo sapiens 10-14 26670903-0 2015 Heme oxygenase-1 attenuates cadmium-induced mitochondrial-caspase 3- dependent apoptosis in human hepatoma cell line. Cadmium 28-35 heme oxygenase 1 Homo sapiens 0-16 26670903-2 2015 In this study, we examined the role of heme oxygenase-1 (HO-1) in modulating the Cd-induced apoptosis in human hepatoma (HepG2) cells after 24 h exposure. Cadmium 81-83 heme oxygenase 1 Homo sapiens 39-55 26670903-2 2015 In this study, we examined the role of heme oxygenase-1 (HO-1) in modulating the Cd-induced apoptosis in human hepatoma (HepG2) cells after 24 h exposure. Cadmium 81-83 heme oxygenase 1 Homo sapiens 57-61 26670903-11 2015 CONCLUSION: In summary, the present study showed that HO-1 attenuates the Cd-induced caspase 3 dependent pathway of apoptosis in HepG2 cells, probably by modulating Cd-induced oxidative stress. Cadmium 165-167 heme oxygenase 1 Homo sapiens 54-58 26655408-6 2015 RESULTS: Oleacein (OC) together with complexes HbHp11 or HbHp22 stimulated the expression of CD163 (30-100-fold), IL-10 (170-300-fold) and HO-1 secretion (60-130-fold) after 5 days of coincubation. oleacein 9-17 heme oxygenase 1 Homo sapiens 139-143 26423448-1 2015 The phenomenon that heme oxygenase-1 (HO-1) protects cell from injury yet its enzymatic product, iron, may facilitate generation of free radical has been long puzzling. Iron 97-101 heme oxygenase 1 Homo sapiens 20-36 26674355-2 2015 Our previous study demonstrated that the flavonoid hyperoside elicits cytoprotection against oxidative stress by activating the Keap1-Nrf2-ARE signaling pathway, thus increasing the expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and catalase. Flavonoids 41-50 heme oxygenase 1 Homo sapiens 225-241 26674355-2 2015 Our previous study demonstrated that the flavonoid hyperoside elicits cytoprotection against oxidative stress by activating the Keap1-Nrf2-ARE signaling pathway, thus increasing the expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and catalase. Flavonoids 41-50 heme oxygenase 1 Homo sapiens 243-247 26674355-2 2015 Our previous study demonstrated that the flavonoid hyperoside elicits cytoprotection against oxidative stress by activating the Keap1-Nrf2-ARE signaling pathway, thus increasing the expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and catalase. hyperoside 51-61 heme oxygenase 1 Homo sapiens 225-241 26674355-2 2015 Our previous study demonstrated that the flavonoid hyperoside elicits cytoprotection against oxidative stress by activating the Keap1-Nrf2-ARE signaling pathway, thus increasing the expression of antioxidant enzymes, such as heme oxygenase-1 (HO-1), superoxide dismutase (SOD) and catalase. hyperoside 51-61 heme oxygenase 1 Homo sapiens 243-247 26674355-6 2015 Further study showed that LiCl and siRNA-mediated inhibition of GSK-3beta increased hyperoside-induced HO-1 expression, and the effect was dependent upon enhanced Nrf2 nuclear translocation and gene expression. hyperoside 84-94 heme oxygenase 1 Homo sapiens 103-107 26674355-8 2015 Furthermore, the siRNA-mediated inhibition of Keap1 also enhanced hyperoside-induced Nrf2 nuclear accumulation and HO-1 expression, which was relatively smaller than the effects obtained from GSK-3beta siRNA administration. hyperoside 66-76 heme oxygenase 1 Homo sapiens 115-119 26674585-0 2015 Effect of oral N-acetylcysteine on COPD patients with microsatellite polymorphism in the heme oxygenase-1 gene promoter. Acetylcysteine 15-31 heme oxygenase 1 Homo sapiens 89-105 26783548-5 2016 RESULTS: Monocytes from multiple sclerosis (MS) patients receiving DMF had reduced expression of the proinflammatory micro-RNA miR-155 and of antioxidant genes HMOX1 and OSGIN1 compared to untreated MS patients; similar changes were observed in patients receiving FTY720 and/or natalizumab. Dimethyl Fumarate 67-70 heme oxygenase 1 Homo sapiens 160-165 26783548-6 2016 In vitro addition of DMF but not MMF to MDMs and microglia inhibited lipopolysaccharide-induced production of inflammatory cytokines and increased expression of the antioxidant gene HMOX1 in the absence of significant cytotoxicity. Dimethyl Fumarate 21-24 heme oxygenase 1 Homo sapiens 182-187 26163109-6 2015 Moreover, the HO-1 inducers, resveratrol and piceatannol decrease the expression of miR-183, resulting in attenuated osteoclastogenesis. 3,3',4,5'-tetrahydroxystilbene 45-56 heme oxygenase 1 Homo sapiens 14-18 26423448-1 2015 The phenomenon that heme oxygenase-1 (HO-1) protects cell from injury yet its enzymatic product, iron, may facilitate generation of free radical has been long puzzling. Iron 97-101 heme oxygenase 1 Homo sapiens 38-42 26423448-6 2015 RNA interference of HO-1 makes the cell more susceptible to hydrogen peroxide, which can be rescued by forced expression of wild-type FHC but not mutants that lose the capacity of iron storage and ferroxidase activity. Hydrogen Peroxide 60-77 heme oxygenase 1 Homo sapiens 20-24 26423448-6 2015 RNA interference of HO-1 makes the cell more susceptible to hydrogen peroxide, which can be rescued by forced expression of wild-type FHC but not mutants that lose the capacity of iron storage and ferroxidase activity. Iron 180-184 heme oxygenase 1 Homo sapiens 20-24 26670322-7 2015 In addition, the induction of HO-1 was also confirmed in normal fibroblasts following incubation with 10-100 micromol/L NAC for 4 h. RESULTS: Following NAC therapy we observed an increase in expression of the antioxidants HO-1 (~4-fold) and its effector ferritin (~160-fold) in patient samples as compared with baseline. Acetylcysteine 120-123 heme oxygenase 1 Homo sapiens 30-34 26670322-7 2015 In addition, the induction of HO-1 was also confirmed in normal fibroblasts following incubation with 10-100 micromol/L NAC for 4 h. RESULTS: Following NAC therapy we observed an increase in expression of the antioxidants HO-1 (~4-fold) and its effector ferritin (~160-fold) in patient samples as compared with baseline. Acetylcysteine 120-123 heme oxygenase 1 Homo sapiens 222-226 26670322-7 2015 In addition, the induction of HO-1 was also confirmed in normal fibroblasts following incubation with 10-100 micromol/L NAC for 4 h. RESULTS: Following NAC therapy we observed an increase in expression of the antioxidants HO-1 (~4-fold) and its effector ferritin (~160-fold) in patient samples as compared with baseline. Acetylcysteine 152-155 heme oxygenase 1 Homo sapiens 30-34 26670322-7 2015 In addition, the induction of HO-1 was also confirmed in normal fibroblasts following incubation with 10-100 micromol/L NAC for 4 h. RESULTS: Following NAC therapy we observed an increase in expression of the antioxidants HO-1 (~4-fold) and its effector ferritin (~160-fold) in patient samples as compared with baseline. Acetylcysteine 152-155 heme oxygenase 1 Homo sapiens 222-226 26087413-9 2015 HO-1 inhibitor and Nrf2 siRNA blocked the milk-induced inflammatory response such as production of ROS, expression of cytokines, chemokines, and MMPs. Reactive Oxygen Species 99-102 heme oxygenase 1 Homo sapiens 0-4 26670322-8 2015 We also observed that NAC exposure significantly increased HO-1 expression in fibroblasts. Acetylcysteine 22-25 heme oxygenase 1 Homo sapiens 59-63 26670322-9 2015 CONCLUSION: Our data suggest that HO-1 is a possible target protein of NAC and a mediator of its cytoprotective effects in these patients. Acetylcysteine 71-74 heme oxygenase 1 Homo sapiens 34-38 26461179-1 2015 Heme-oxygenase 1 (HO-1) prevents T cell-mediated inflammatory disease by producing carbon monoxide (CO) and impairing DC immunogenicity. Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 0-16 26461179-1 2015 Heme-oxygenase 1 (HO-1) prevents T cell-mediated inflammatory disease by producing carbon monoxide (CO) and impairing DC immunogenicity. Carbon Monoxide 100-102 heme oxygenase 1 Homo sapiens 0-16 26456836-11 2015 Curcumin also inhibited superoxide anion-induced leukocyte recruitment in the peritoneal cavity and in the paw skin inhibited myeloperoxidase activity, oxidative stress, IL-1beta and TNF-alpha production and NF-kappaB activation as well as enhanced IL-10 production, and HO-1 and Nrf2 mRNA expression. Curcumin 0-8 heme oxygenase 1 Homo sapiens 271-275 26315932-2 2015 We have previously shown that the liberation of heme in acute hemolysis can induce heme oxygenase-1 during granulopoiesis, impairing the ability of developing neutrophils to mount a bactericidal oxidative burst, and increasing susceptibility to bacterial infection. Heme 48-52 heme oxygenase 1 Homo sapiens 83-99 26456060-12 2015 PAMPs such as bacterial endotoxin activate HO-1, and the CO that is generated diffuses into the extracellular milieu where it interacts with bacteria, altering their behavior to increase production of ATP, which then functions as a second signal danger molecule. Adenosine Triphosphate 201-204 heme oxygenase 1 Homo sapiens 43-47 26507166-4 2015 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes oxidation of heme to biliverdin, and has anti-inflammatory and anti-oxidant properties. Heme 65-69 heme oxygenase 1 Homo sapiens 0-16 26456836-11 2015 Curcumin also inhibited superoxide anion-induced leukocyte recruitment in the peritoneal cavity and in the paw skin inhibited myeloperoxidase activity, oxidative stress, IL-1beta and TNF-alpha production and NF-kappaB activation as well as enhanced IL-10 production, and HO-1 and Nrf2 mRNA expression. Superoxides 24-40 heme oxygenase 1 Homo sapiens 271-275 26507166-4 2015 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes oxidation of heme to biliverdin, and has anti-inflammatory and anti-oxidant properties. Heme 65-69 heme oxygenase 1 Homo sapiens 18-22 26507166-4 2015 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes oxidation of heme to biliverdin, and has anti-inflammatory and anti-oxidant properties. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 0-16 26507166-4 2015 Heme oxygenase-1 (HO-1) is an enzyme that catalyzes oxidation of heme to biliverdin, and has anti-inflammatory and anti-oxidant properties. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 18-22 26507166-11 2015 IL-13 significantly increased goblet cells, MUC5AC protein secretion (p b 0.01) and MUC5AC mRNA (p b 0.001), and these were decreased by hemin by way of HO-1. Hemin 137-142 heme oxygenase 1 Homo sapiens 153-157 26507166-12 2015 Tin protoporphyrin (SnPP)-IX, a HO-1 inhibitor, blocked the effect of hemin restoring MUC5AC protein secretion (p b 0.05) and goblet cell hyperplasia. tin protoporphyrin IX 0-18 heme oxygenase 1 Homo sapiens 32-36 26507166-12 2015 Tin protoporphyrin (SnPP)-IX, a HO-1 inhibitor, blocked the effect of hemin restoring MUC5AC protein secretion (p b 0.05) and goblet cell hyperplasia. S-Nitroso-N-propionyl-D,L-penicillamine 20-24 heme oxygenase 1 Homo sapiens 32-36 26590114-0 2015 Fisetin inhibits TNF-alpha-induced inflammatory action and hydrogen peroxide-induced oxidative damage in human keratinocyte HaCaT cells through PI3K/AKT/Nrf-2-mediated heme oxygenase-1 expression. fisetin 0-7 heme oxygenase 1 Homo sapiens 168-184 26590114-5 2015 When HaCaT cells were treated with non-cytotoxic concentrations of fisetin (1-20muM), heme oxygenase (HO)-1 mRNA and protein expression increased in a dose-dependent manner. fisetin 67-74 heme oxygenase 1 Homo sapiens 86-107 26431832-0 2015 Guanosine inhibits LPS-induced pro-inflammatory response and oxidative stress in hippocampal astrocytes through the heme oxygenase-1 pathway. Guanosine 0-9 heme oxygenase 1 Homo sapiens 116-132 26240115-4 2015 Olmesartan, an Ang II type 1 receptor blocker, possesses antioxidant and activating nitric oxide system-related effects, which we have shown in terms of p22(phox) reduction, heme oxygenase-1 and calcitonin gene-related peptide increase. olmesartan 0-10 heme oxygenase 1 Homo sapiens 174-190 26432957-5 2015 HO-1 degrades heme to biliverdin, carbon monoxide (CO) and free iron. Biliverdine 22-32 heme oxygenase 1 Homo sapiens 0-4 26432957-5 2015 HO-1 degrades heme to biliverdin, carbon monoxide (CO) and free iron. Carbon Monoxide 34-49 heme oxygenase 1 Homo sapiens 0-4 26432957-5 2015 HO-1 degrades heme to biliverdin, carbon monoxide (CO) and free iron. Carbon Monoxide 51-53 heme oxygenase 1 Homo sapiens 0-4 26432957-5 2015 HO-1 degrades heme to biliverdin, carbon monoxide (CO) and free iron. Iron 64-68 heme oxygenase 1 Homo sapiens 0-4 26432957-6 2015 Biliverdin and CO are gaining particular interest because these two have been found to mediate most of anti-inflammatory, antioxidant and anti-apoptotic effects of HO-1. Biliverdine 0-10 heme oxygenase 1 Homo sapiens 164-168 26432957-6 2015 Biliverdin and CO are gaining particular interest because these two have been found to mediate most of anti-inflammatory, antioxidant and anti-apoptotic effects of HO-1. Carbon Monoxide 15-17 heme oxygenase 1 Homo sapiens 164-168 26431832-2 2015 Our group recently demonstrated that guanosine exhibits glioprotective effects in the C6 astroglial cell line by associating the heme oxygenase-1 (HO-1) signaling pathway with protection against azide-induced oxidative stress. Guanosine 37-46 heme oxygenase 1 Homo sapiens 129-145 26431832-2 2015 Our group recently demonstrated that guanosine exhibits glioprotective effects in the C6 astroglial cell line by associating the heme oxygenase-1 (HO-1) signaling pathway with protection against azide-induced oxidative stress. Guanosine 37-46 heme oxygenase 1 Homo sapiens 147-151 26431832-2 2015 Our group recently demonstrated that guanosine exhibits glioprotective effects in the C6 astroglial cell line by associating the heme oxygenase-1 (HO-1) signaling pathway with protection against azide-induced oxidative stress. Azides 195-200 heme oxygenase 1 Homo sapiens 129-145 26431832-2 2015 Our group recently demonstrated that guanosine exhibits glioprotective effects in the C6 astroglial cell line by associating the heme oxygenase-1 (HO-1) signaling pathway with protection against azide-induced oxidative stress. Azides 195-200 heme oxygenase 1 Homo sapiens 147-151 26431832-8 2015 Guanosine was able to prevent these effects, protecting the hippocampal astrocytes against LPS-induced cytotoxicity through activation of the HO-1 pathway. Guanosine 0-9 heme oxygenase 1 Homo sapiens 142-146 26414010-8 2015 Alpha-lipoic acid, an oxidative stress inhibitor, prevented the development of superoxide acutely and increased antiapoptotic markers B-cell lymphoma 2 (t = 3.079, P < 0.05) and heme oxygenase 1 (t = 8.169, P < 0.001) after single blast. Thioctic Acid 0-17 heme oxygenase 1 Homo sapiens 181-197 26299779-2 2015 We have previously demonstrated that preconditioning human CSCs (hCSCs) with the heme oxygenase-1 inducer, cobalt protoporphyrin (CoPP), has significant cytoprotective effects in vitro. cobaltiprotoporphyrin 107-128 heme oxygenase 1 Homo sapiens 81-97 26299779-2 2015 We have previously demonstrated that preconditioning human CSCs (hCSCs) with the heme oxygenase-1 inducer, cobalt protoporphyrin (CoPP), has significant cytoprotective effects in vitro. cobaltiprotoporphyrin 130-134 heme oxygenase 1 Homo sapiens 81-97 26385185-1 2015 Carbon monoxide (CO) is one of the cytoprotective byproducts of heme oxygenase (HO)-1 and exerts anti-inflammatory action in various models. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 64-85 26385185-1 2015 Carbon monoxide (CO) is one of the cytoprotective byproducts of heme oxygenase (HO)-1 and exerts anti-inflammatory action in various models. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 64-85 26318273-8 2015 Moreover, we demonstrated that heme oxygenase 1 (HO1), an enzyme that is part of the cellular defense system, mediated some of the effects of resveratrol. Resveratrol 142-153 heme oxygenase 1 Homo sapiens 31-47 26318273-8 2015 Moreover, we demonstrated that heme oxygenase 1 (HO1), an enzyme that is part of the cellular defense system, mediated some of the effects of resveratrol. Resveratrol 142-153 heme oxygenase 1 Homo sapiens 49-52 26870816-3 2016 To limit the deleterious effects of excess ROS, certain gene promoters contain antioxidant response elements (ARE), e.g. the genes NQO-1 and HO-1. Reactive Oxygen Species 43-46 heme oxygenase 1 Homo sapiens 141-145 26611539-0 2015 Brazilian green propolis water extract up-regulates the early expression level of HO-1 and accelerates Nrf2 after UVA irradiation. Water 25-30 heme oxygenase 1 Homo sapiens 82-86 26600472-7 2015 We found that mercury increases oxidative stress (increased HO1 and NQO1 mRNA levels) and alters the cytoskeleton in the human endometrial Ishikawa cell line and to a lesser extent, in the "less-differentiated" human endometrial Hec-1b cells. Mercury 14-21 heme oxygenase 1 Homo sapiens 60-63 26556982-7 2015 Although the underlying mechanisms are not well defined for most of the antibiotics, the pathways triggered for norfloxacin to induce such immunomodulatory effects involve the down-regulation of pro-inflammatory inducible nitric oxide synthase, cyclooxygenase-2, and NF-kappaB and the up-regulation of heme-oxygenase 1 and IL-10 expression. Norfloxacin 112-123 heme oxygenase 1 Homo sapiens 302-318 26472707-8 2015 Results indicate that Cucurbitacins markedly promoted the activation of Nrf-2/ARE pathway-related downstream factors including NQO-1 and HO-1. Cucurbitacins 22-35 heme oxygenase 1 Homo sapiens 137-141 26472707-9 2015 Furthermore, anti-neuroinflammatory effects of Cucurbitacins are attenuated in the knockdown of Nrf2, HO-1 and NQO-1 respectively. Cucurbitacins 47-60 heme oxygenase 1 Homo sapiens 102-106 26884897-8 2015 HO-1 and its reaction products of heme degradation has been linked to cytoprotection, and as an inducible form of HO, serves a vital metabolic function as the rate-limiting step in the heme degradation pathway, and affords protection in models of liver I/R injury. Heme 34-38 heme oxygenase 1 Homo sapiens 0-4 26884897-8 2015 HO-1 and its reaction products of heme degradation has been linked to cytoprotection, and as an inducible form of HO, serves a vital metabolic function as the rate-limiting step in the heme degradation pathway, and affords protection in models of liver I/R injury. Heme 185-189 heme oxygenase 1 Homo sapiens 0-4 25847254-5 2015 In addition, 10-MDP induced activation of nuclear factor-E2-related factor 2 (Nrf2) and its target gene, haeme oxygenase-1 (HO-1). methacryloyloxydecyl dihydrogen phosphate 13-19 heme oxygenase 1 Homo sapiens 105-122 25847254-5 2015 In addition, 10-MDP induced activation of nuclear factor-E2-related factor 2 (Nrf2) and its target gene, haeme oxygenase-1 (HO-1). methacryloyloxydecyl dihydrogen phosphate 13-19 heme oxygenase 1 Homo sapiens 124-128 25847254-6 2015 We evaluated whether the effect of 10-MDP was related to the induction of HO-1 and found that treatment with a selective inhibitor of HO-1 reversed the production of 10-MDP-mediated pro-inflammatory cytokines and the inhibition of differentiation markers. methacryloyloxydecyl dihydrogen phosphate 35-41 heme oxygenase 1 Homo sapiens 74-78 25847254-6 2015 We evaluated whether the effect of 10-MDP was related to the induction of HO-1 and found that treatment with a selective inhibitor of HO-1 reversed the production of 10-MDP-mediated pro-inflammatory cytokines and the inhibition of differentiation markers. methacryloyloxydecyl dihydrogen phosphate 35-41 heme oxygenase 1 Homo sapiens 134-138 25847254-6 2015 We evaluated whether the effect of 10-MDP was related to the induction of HO-1 and found that treatment with a selective inhibitor of HO-1 reversed the production of 10-MDP-mediated pro-inflammatory cytokines and the inhibition of differentiation markers. methacryloyloxydecyl dihydrogen phosphate 166-172 heme oxygenase 1 Homo sapiens 74-78 25847254-6 2015 We evaluated whether the effect of 10-MDP was related to the induction of HO-1 and found that treatment with a selective inhibitor of HO-1 reversed the production of 10-MDP-mediated pro-inflammatory cytokines and the inhibition of differentiation markers. methacryloyloxydecyl dihydrogen phosphate 166-172 heme oxygenase 1 Homo sapiens 134-138 26351029-0 2015 Organic Nitrates in Heart Failure Revisited: Pentaerythritol Tetranitrate Induces Heme Oxygenase 1 to Protect the Myocardium. organic nitrates 0-16 heme oxygenase 1 Homo sapiens 82-98 26456587-8 2015 Furthermore, SMXZF not only dose-dependently inhibited the phosphorylation of NF-kappaB, p50, p65 and IKKalpha/beta in TNF-alpha-treated EA.hy926 cells, but also regulated the Nrf2/HO-1 pathway in H2O2-treated PC12 cells. smxzf 13-18 heme oxygenase 1 Homo sapiens 181-185 26324507-8 2015 Administering tin protoporphyrin IX dichloride (HO-1 inhibitor) or cobalt protoporphyrin (HO-1 inducer) into placental explants did not affect sFlt-1 or sENG secretion. cobaltiprotoporphyrin 67-88 heme oxygenase 1 Homo sapiens 90-94 26351029-0 2015 Organic Nitrates in Heart Failure Revisited: Pentaerythritol Tetranitrate Induces Heme Oxygenase 1 to Protect the Myocardium. Pentaerythritol Tetranitrate 45-73 heme oxygenase 1 Homo sapiens 82-98 26722274-0 2015 Knockdown of heme oxygenase-1 promotes apoptosis and autophagy and enhances the cytotoxicity of doxorubicin in breast cancer cells. Doxorubicin 96-107 heme oxygenase 1 Homo sapiens 13-29 26227317-9 2015 Induction of DNA damage by mitoxantrone and etoposide repressed HO-1 transcription, whereas hydrogen peroxide and doxorubicin induced its expression. Mitoxantrone 27-39 heme oxygenase 1 Homo sapiens 64-68 26227317-9 2015 Induction of DNA damage by mitoxantrone and etoposide repressed HO-1 transcription, whereas hydrogen peroxide and doxorubicin induced its expression. Etoposide 44-53 heme oxygenase 1 Homo sapiens 64-68 26227317-10 2015 Xenograft studies showed that HO-1 regulation by doxorubicin also occurs in vivo. Doxorubicin 49-60 heme oxygenase 1 Homo sapiens 30-34 26227317-11 2015 Immunofluorescence analysis revealed that BRCA1 overexpression and/or doxorubicin exposure induced the cytoplasmic retention of HO-1. Doxorubicin 70-81 heme oxygenase 1 Homo sapiens 128-132 26722301-5 2015 In the present study, HO-1 was expressed in the human AML Kasumi-1, HL-60 and THP-1 cell lines, and HO-1 expression was regulated by Hemin (20 micromol/l) and ZnPPIX (10 micromol/l). zinc protoporphyrin 159-165 heme oxygenase 1 Homo sapiens 22-26 26722301-5 2015 In the present study, HO-1 was expressed in the human AML Kasumi-1, HL-60 and THP-1 cell lines, and HO-1 expression was regulated by Hemin (20 micromol/l) and ZnPPIX (10 micromol/l). zinc protoporphyrin 159-165 heme oxygenase 1 Homo sapiens 100-104 26722274-2 2015 In the present study, HMOX-1 was demonstrated to be overexpressed and able to be induced by doxorubicin in breast cancer cell lines. Doxorubicin 92-103 heme oxygenase 1 Homo sapiens 22-28 26722274-3 2015 Knockdown of HMOX-1 using short interfering (si)RNA enhanced the cytotoxicity of doxorubicin in MDA-MB-231 and BT549 cells. Doxorubicin 81-92 heme oxygenase 1 Homo sapiens 13-19 26722274-4 2015 Knockdown of HMOX-1 downregulated B cell lymphoma (Bcl)-2 and Bcl-extra large expression, and significantly enhanced doxorubicin-induced apoptosis in MDA-MB-231 and BT549 cells. Doxorubicin 117-128 heme oxygenase 1 Homo sapiens 13-19 26722274-5 2015 Additionally, knockdown of HMOX-1 upregulated light chain 3B expression and markedly increased the accumulation of autophagic vacuoles in MDA-MB-231 and BT549 cells treated with doxorubicin. Doxorubicin 178-189 heme oxygenase 1 Homo sapiens 27-33 26722274-7 2015 Therefore, HMOX-1 may represent a potential therapeutic target for enhancing the cytotoxicity and efficacy of doxorubicin during the treatment of breast cancer. Doxorubicin 110-121 heme oxygenase 1 Homo sapiens 11-17 26147741-1 2015 Heme Oxygenase-1 and its product biliverdin/bilirubin have been demonstrated to protect against ischemia/reperfusion injury (IRI). Biliverdine 33-43 heme oxygenase 1 Homo sapiens 0-16 26392237-1 2015 Heme oxygenase-1 (HO-1, hmox-1) catalyzes the rate-limiting step in the heme degradation processes. Heme 72-76 heme oxygenase 1 Homo sapiens 0-22 26392237-1 2015 Heme oxygenase-1 (HO-1, hmox-1) catalyzes the rate-limiting step in the heme degradation processes. Heme 72-76 heme oxygenase 1 Homo sapiens 24-30 26392237-2 2015 Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Biliverdine 43-53 heme oxygenase 1 Homo sapiens 146-150 26392237-2 2015 Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Iron 55-59 heme oxygenase 1 Homo sapiens 146-150 26392237-2 2015 Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 146-150 26392237-2 2015 Out of three by-products of HO-1 activity, biliverdin, iron ions and carbon monoxide (CO), the latter was mostly shown to mediate many beneficial HO-1 effects, including protection against oxidative injury, regulation of apoptosis, modulation of inflammation as well as contribution to angiogenesis. Carbon Monoxide 86-88 heme oxygenase 1 Homo sapiens 146-150 26418896-5 2015 Application of CO, a product of HO-1 catalysis, increased levels of E-cadherin in the adherens junctions between cancer cells. Carbon Monoxide 15-17 heme oxygenase 1 Homo sapiens 32-36 26510767-2 2015 Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Heme 112-116 heme oxygenase 1 Homo sapiens 0-16 26510767-2 2015 Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Heme 112-116 heme oxygenase 1 Homo sapiens 18-22 26510767-7 2015 Increasing HO-1 expression through adenoviral-mediated overexpression or induction with cobalt protoporphyrin (CoPP, a potent HO-1 inducer), pre- and postinfection, effectively inhibited BVDV replication. cobaltiprotoporphyrin 88-109 heme oxygenase 1 Homo sapiens 11-15 26510767-7 2015 Increasing HO-1 expression through adenoviral-mediated overexpression or induction with cobalt protoporphyrin (CoPP, a potent HO-1 inducer), pre- and postinfection, effectively inhibited BVDV replication. cobaltiprotoporphyrin 88-109 heme oxygenase 1 Homo sapiens 126-130 26510767-7 2015 Increasing HO-1 expression through adenoviral-mediated overexpression or induction with cobalt protoporphyrin (CoPP, a potent HO-1 inducer), pre- and postinfection, effectively inhibited BVDV replication. cobaltiprotoporphyrin 111-115 heme oxygenase 1 Homo sapiens 11-15 26510767-7 2015 Increasing HO-1 expression through adenoviral-mediated overexpression or induction with cobalt protoporphyrin (CoPP, a potent HO-1 inducer), pre- and postinfection, effectively inhibited BVDV replication. cobaltiprotoporphyrin 111-115 heme oxygenase 1 Homo sapiens 126-130 26186946-1 2015 The heme oxygenase-1 (HO-1)/carbon monoxide (CO) system induces mitochondrial biogenesis, but its biological impact in human skeletal muscle is uncertain. Carbon Monoxide 45-47 heme oxygenase 1 Homo sapiens 4-20 26186946-1 2015 The heme oxygenase-1 (HO-1)/carbon monoxide (CO) system induces mitochondrial biogenesis, but its biological impact in human skeletal muscle is uncertain. Carbon Monoxide 45-47 heme oxygenase 1 Homo sapiens 22-26 26493719-3 2015 Heme oxygenase-1 (HO-1; also known as Hsp32) is an inducible enzyme participating in heme degradation and involved in oxidative stress resistance. Heme 85-89 heme oxygenase 1 Homo sapiens 0-16 26350099-5 2015 Furthermore, with the addition of N-acetylcysteine, a scavenger of reactive oxygen species (ROS), it was found that NGAL depletion was sufficient to cause apoptosis of lung adenocarcinoma cells by generating ROS through the inhibition of the nuclear factor E2-related factor 2/heme oxygenase-1 anti-oxidant pathway. Acetylcysteine 34-50 heme oxygenase 1 Homo sapiens 277-293 26341012-12 2015 Plumbagin increased TrxR-1 and heme oxygenase (HO)-1 expression and pretreatment with NAC significantly attenuated the plumbagin-induced increase of TrxR-1 and HO-1 expression in HepG2 cells, LLC cells and SiHa cells. Acetylcysteine 86-89 heme oxygenase 1 Homo sapiens 160-164 26493719-3 2015 Heme oxygenase-1 (HO-1; also known as Hsp32) is an inducible enzyme participating in heme degradation and involved in oxidative stress resistance. Heme 85-89 heme oxygenase 1 Homo sapiens 18-22 26493719-3 2015 Heme oxygenase-1 (HO-1; also known as Hsp32) is an inducible enzyme participating in heme degradation and involved in oxidative stress resistance. Heme 85-89 heme oxygenase 1 Homo sapiens 38-43 26291278-0 2015 Coordinated regulation of Nrf2 and histone H3 serine 10 phosphorylation in arsenite-activated transcription of the human heme oxygenase-1 gene. Serine 46-52 heme oxygenase 1 Homo sapiens 121-137 26291278-8 2015 In response to arsenite, binding of Nrf2 to the HO-1 ARE preceded phosphorylation of H3S10 at the HO-1 ARE. arsenite 15-23 heme oxygenase 1 Homo sapiens 48-52 26291278-0 2015 Coordinated regulation of Nrf2 and histone H3 serine 10 phosphorylation in arsenite-activated transcription of the human heme oxygenase-1 gene. arsenite 75-83 heme oxygenase 1 Homo sapiens 121-137 26291278-8 2015 In response to arsenite, binding of Nrf2 to the HO-1 ARE preceded phosphorylation of H3S10 at the HO-1 ARE. arsenite 15-23 heme oxygenase 1 Homo sapiens 98-102 26291278-4 2015 The environmental contaminant arsenite is a potent inducer of both HO-1 expression and phosphorylation of histone H3 serine 10 (H3S10); therefore, we investigated the relationships between Nrf2 and H3S10 phosphorylation in arsenite-induced, ARE-dependent, transcriptional activation of the human HO-1 gene. arsenite 30-38 heme oxygenase 1 Homo sapiens 67-71 26291278-4 2015 The environmental contaminant arsenite is a potent inducer of both HO-1 expression and phosphorylation of histone H3 serine 10 (H3S10); therefore, we investigated the relationships between Nrf2 and H3S10 phosphorylation in arsenite-induced, ARE-dependent, transcriptional activation of the human HO-1 gene. arsenite 30-38 heme oxygenase 1 Homo sapiens 296-300 26291278-8 2015 In response to arsenite, binding of Nrf2 to the HO-1 ARE preceded phosphorylation of H3S10 at the HO-1 ARE. h3s10 85-90 heme oxygenase 1 Homo sapiens 48-52 26291278-8 2015 In response to arsenite, binding of Nrf2 to the HO-1 ARE preceded phosphorylation of H3S10 at the HO-1 ARE. h3s10 85-90 heme oxygenase 1 Homo sapiens 98-102 26291278-5 2015 Arsenite increased phosphorylation of H3S10 both globally and at the HO-1 promoter concomitantly with HO-1 transcription in human HaCaT keratinocytes. arsenite 0-8 heme oxygenase 1 Homo sapiens 69-73 26291278-9 2015 Furthermore, arsenite-mediated occupancy of phosphorylated H3S10 at the HO-1 ARE was decreased in Nrf2-deficient mouse embryonic fibroblasts. arsenite 13-21 heme oxygenase 1 Homo sapiens 72-76 26291278-5 2015 Arsenite increased phosphorylation of H3S10 both globally and at the HO-1 promoter concomitantly with HO-1 transcription in human HaCaT keratinocytes. h3s10 38-43 heme oxygenase 1 Homo sapiens 69-73 26291278-9 2015 Furthermore, arsenite-mediated occupancy of phosphorylated H3S10 at the HO-1 ARE was decreased in Nrf2-deficient mouse embryonic fibroblasts. h3s10 59-64 heme oxygenase 1 Homo sapiens 72-76 26291278-10 2015 These results suggest the involvement of H3S10 phosphorylation in the Nrf2-ARE axis by proposing that Nrf2 may influence H3S10 phosphorylation at the HO-1 ARE and additional promoter regions. h3s10 41-46 heme oxygenase 1 Homo sapiens 150-154 26291278-6 2015 Conversely, arsenite-induced H3S10 phosphorylation and HO-1 expression were blocked by N-acetylcysteine (NAC), the c-Jun N-terminal kinase (JNK) inhibitor SP600125, and JNK knockdown (siJNK). arsenite 12-20 heme oxygenase 1 Homo sapiens 55-59 26291278-10 2015 These results suggest the involvement of H3S10 phosphorylation in the Nrf2-ARE axis by proposing that Nrf2 may influence H3S10 phosphorylation at the HO-1 ARE and additional promoter regions. h3s10 121-126 heme oxygenase 1 Homo sapiens 150-154 26291278-11 2015 Our data highlights the complex interplay between Nrf2 and H3S10 phosphorylation in arsenite-activated HO-1 transcription. h3s10 59-64 heme oxygenase 1 Homo sapiens 103-107 26291278-6 2015 Conversely, arsenite-induced H3S10 phosphorylation and HO-1 expression were blocked by N-acetylcysteine (NAC), the c-Jun N-terminal kinase (JNK) inhibitor SP600125, and JNK knockdown (siJNK). Acetylcysteine 87-103 heme oxygenase 1 Homo sapiens 55-59 26291278-11 2015 Our data highlights the complex interplay between Nrf2 and H3S10 phosphorylation in arsenite-activated HO-1 transcription. arsenite 84-92 heme oxygenase 1 Homo sapiens 103-107 26291278-6 2015 Conversely, arsenite-induced H3S10 phosphorylation and HO-1 expression were blocked by N-acetylcysteine (NAC), the c-Jun N-terminal kinase (JNK) inhibitor SP600125, and JNK knockdown (siJNK). Acetylcysteine 105-108 heme oxygenase 1 Homo sapiens 55-59 26416516-8 2015 Terrein induced the induction of anti-oxidant molecules, copper/zinc-superoxide defence (Cu/ZnSOD), manganese superoxide dismutase (MnSOD) and heme oxygenase-1 (HO-1) in SIPS cells. terrein 0-7 heme oxygenase 1 Homo sapiens 143-159 26416516-8 2015 Terrein induced the induction of anti-oxidant molecules, copper/zinc-superoxide defence (Cu/ZnSOD), manganese superoxide dismutase (MnSOD) and heme oxygenase-1 (HO-1) in SIPS cells. terrein 0-7 heme oxygenase 1 Homo sapiens 161-165 26436888-8 2015 RESULTS: Treatment with 30 or 50 muM resveratrol reduced ROS production and HO-1 level induced by oxidative stress. Resveratrol 37-48 heme oxygenase 1 Homo sapiens 76-80 26339767-5 2015 RECENT FINDINGS: Recent studies demonstrate that Hx-dependent uptake of extracellular heme leads to the deactivation of Bach1 repression leading to the transcriptional activation of antioxidant heme oxygenase-1 gene. Heme 86-90 heme oxygenase 1 Homo sapiens 194-210 26206798-8 2015 HMOX1 rs743811 associated with chronic kidney disease stage (OR=3.0, P=0.0001) in the University of Illinois cohort and end-stage renal disease (OR=10.0, P=0.0003) in the Walk-Treatment of Pulmonary Hypertension and Sickle cell Disease with Sildenafil Therapy cohort. Sildenafil Citrate 241-251 heme oxygenase 1 Homo sapiens 0-5 26043815-0 2015 Lipoic acid and N-acetylcysteine prevent ammonia-induced inflammatory response in C6 astroglial cells: The putative role of ERK and HO1 signaling pathways. Thioctic Acid 0-11 heme oxygenase 1 Homo sapiens 132-135 25921464-5 2015 We showed that CORM-2-induced HO-1 protein and mRNA levels were inhibited by the inhibitor of c-Src (PP1 or SU6656), EGFR (AG1478), PI3K (LY294002), Akt (SH-5), JNK1/2 (SP600125), or p38 MAPK (SB202190) and transfection with siRNA of c-Src, EGFR, Akt, p38, JNK2, or Nrf2 in HTSMCs. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 138-146 heme oxygenase 1 Homo sapiens 30-34 25921464-5 2015 We showed that CORM-2-induced HO-1 protein and mRNA levels were inhibited by the inhibitor of c-Src (PP1 or SU6656), EGFR (AG1478), PI3K (LY294002), Akt (SH-5), JNK1/2 (SP600125), or p38 MAPK (SB202190) and transfection with siRNA of c-Src, EGFR, Akt, p38, JNK2, or Nrf2 in HTSMCs. pyrazolanthrone 169-177 heme oxygenase 1 Homo sapiens 30-34 26141635-7 2015 Under normal conditions, fHb is effectively scavenged by the Hb-scavenging mechanism (HSM); this involves binding to haptoglobin (Hp), uptake via the Hb-scavenging receptor (CD163) on monocytes and metabolism by heme-oxygenase-1. 3-FLUORO-4-HYDROXYBENZOIC ACID 25-28 heme oxygenase 1 Homo sapiens 212-228 26929573-4 2015 MATERIALS AND METHODS: Protective ability of flavonoid-enriched (FE) fraction of clove was studied against UV-B induced cytotoxicity, anti-oxidant regulation, oxidative DNA damage, intracellular reactive oxygen species (ROS) generation, apoptotic morphological changes, and regulation of heme oxygenase-1 (HO-1) gene through nuclear factor E2-related factor 2 antioxidant response element (Nrf2 ARE) pathway. Flavonoids 45-54 heme oxygenase 1 Homo sapiens 306-310 26929573-7 2015 CONCLUSION: The present study demonstrated for the first time that the FE fraction from clove could confer UV-B protection probably through the Nrf2-ARE pathway, which included the down-regulation of Nrf2 and HO-1. Iron 71-73 heme oxygenase 1 Homo sapiens 209-213 25921464-1 2015 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, displays an anti-inflammatory effect in various cellular injuries, but the precise mechanisms of HO-1 expression remain unknown. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 63-84 25921464-1 2015 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, displays an anti-inflammatory effect in various cellular injuries, but the precise mechanisms of HO-1 expression remain unknown. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 183-187 25921464-1 2015 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, displays an anti-inflammatory effect in various cellular injuries, but the precise mechanisms of HO-1 expression remain unknown. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 63-84 25921464-1 2015 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, displays an anti-inflammatory effect in various cellular injuries, but the precise mechanisms of HO-1 expression remain unknown. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 183-187 25921464-5 2015 We showed that CORM-2-induced HO-1 protein and mRNA levels were inhibited by the inhibitor of c-Src (PP1 or SU6656), EGFR (AG1478), PI3K (LY294002), Akt (SH-5), JNK1/2 (SP600125), or p38 MAPK (SB202190) and transfection with siRNA of c-Src, EGFR, Akt, p38, JNK2, or Nrf2 in HTSMCs. SU 6656 108-114 heme oxygenase 1 Homo sapiens 30-34 26185204-10 2015 Furthermore, activated ascaridole showed increased potential for induction of the Nrf2 target gene heme oxygenase 1 and upregulation of CD86 and CD54 on THP-1 cells, an established DC surrogate. ascaridole 23-33 heme oxygenase 1 Homo sapiens 99-115 26043815-5 2015 LA and NAC prevented these effects by the modulation of ERK and HO1 pathways. Acetylcysteine 7-10 heme oxygenase 1 Homo sapiens 64-67 26192096-0 2015 Involvement of the Nrf2/HO-1 signaling pathway in sulfuretin-induced protection against amyloid beta25-35 neurotoxicity. sulfuretin 50-60 heme oxygenase 1 Homo sapiens 24-28 26277392-0 2015 Nitration of indoxyl sulfate facilitates its cytotoxicity in human renal proximal tubular cells via expression of heme oxygenase-1. Indican 13-28 heme oxygenase 1 Homo sapiens 114-130 26277392-7 2015 Most importantly, however, as detected by immunofluorescence and Western blotting, 2-nitro-IS induces the expression of heme oxygenase-1 and thereby the formation of ROS; most probably through the Fenton reaction. 2-nitro 83-90 heme oxygenase 1 Homo sapiens 120-136 26277392-7 2015 Most importantly, however, as detected by immunofluorescence and Western blotting, 2-nitro-IS induces the expression of heme oxygenase-1 and thereby the formation of ROS; most probably through the Fenton reaction. Reactive Oxygen Species 166-169 heme oxygenase 1 Homo sapiens 120-136 26192096-6 2015 We demonstrated that sulfuretin induces the expression of heme oxygenase-1 (HO-1), an anti-oxidant response gene. sulfuretin 21-31 heme oxygenase 1 Homo sapiens 58-74 26192096-6 2015 We demonstrated that sulfuretin induces the expression of heme oxygenase-1 (HO-1), an anti-oxidant response gene. sulfuretin 21-31 heme oxygenase 1 Homo sapiens 76-80 26143632-4 2015 Treatment of human adipocytes with cobalt protoporphyrin (CoPP) or hemin for 24-48 h increased HO-1 expression and activity without affecting adiponectin expression and secretion. cobaltiprotoporphyrin 35-56 heme oxygenase 1 Homo sapiens 95-99 26192096-7 2015 Notably, we found that the neuroprotective effects of sulfuretin were diminished by an Nrf2 small interfering RNA (siRNA), the HO-1 inhibitor zinc protoporphyrin IX (ZnPP), as well as the PI3K/Akt inhibitor LY294002. sulfuretin 54-64 heme oxygenase 1 Homo sapiens 127-131 26192096-8 2015 Taken together, these results indicated that sulfuretin protects neuronal cells from Abeta25-35-induced neurotoxicity through activation of Nrf/HO-1 and PI3K/Akt signaling pathways. sulfuretin 45-55 heme oxygenase 1 Homo sapiens 144-148 26192096-9 2015 Our results also indicate that sulfuretin-induced induction of Nrf2-dependent HO-1 expression via the PI3K/Akt signaling pathway has preventive and/or therapeutic potential for the management of Alzheimer"s disease. sulfuretin 31-41 heme oxygenase 1 Homo sapiens 78-82 26275128-0 2015 Novel Nrf2/ARE activator, trans-Coniferylaldehyde, induces a HO-1-mediated defense mechanism through a dual p38alpha/MAPKAPK-2 and PK-N3 signaling pathway. coniferaldehyde 26-49 heme oxygenase 1 Homo sapiens 61-65 26275128-6 2015 The detoxifying/antioxidant genes activated by t-CA, especially heme oxygenase-1 (HO-1), were found to be involved in its cytoprotective effects against oxidative stress and cell injuries elicited by carcinogens tert-butylhydroperoxide and arecoline. tert-Butylhydroperoxide 212-235 heme oxygenase 1 Homo sapiens 64-80 26275128-6 2015 The detoxifying/antioxidant genes activated by t-CA, especially heme oxygenase-1 (HO-1), were found to be involved in its cytoprotective effects against oxidative stress and cell injuries elicited by carcinogens tert-butylhydroperoxide and arecoline. tert-Butylhydroperoxide 212-235 heme oxygenase 1 Homo sapiens 82-86 26275128-6 2015 The detoxifying/antioxidant genes activated by t-CA, especially heme oxygenase-1 (HO-1), were found to be involved in its cytoprotective effects against oxidative stress and cell injuries elicited by carcinogens tert-butylhydroperoxide and arecoline. Arecoline 240-249 heme oxygenase 1 Homo sapiens 64-80 26275128-6 2015 The detoxifying/antioxidant genes activated by t-CA, especially heme oxygenase-1 (HO-1), were found to be involved in its cytoprotective effects against oxidative stress and cell injuries elicited by carcinogens tert-butylhydroperoxide and arecoline. Arecoline 240-249 heme oxygenase 1 Homo sapiens 82-86 26445536-10 2015 In summary, AuNPs enhance the levels and nuclear translocation of the Nrf2 protein and Bach1 export/tyrosine phosphorylation, leading to Nrf2 binding to the HO-1 E2 enhancer promoter region to drive HO-1 expression in ECs. Tyrosine 100-108 heme oxygenase 1 Homo sapiens 157-161 26445536-10 2015 In summary, AuNPs enhance the levels and nuclear translocation of the Nrf2 protein and Bach1 export/tyrosine phosphorylation, leading to Nrf2 binding to the HO-1 E2 enhancer promoter region to drive HO-1 expression in ECs. Tyrosine 100-108 heme oxygenase 1 Homo sapiens 199-203 26143632-4 2015 Treatment of human adipocytes with cobalt protoporphyrin (CoPP) or hemin for 24-48 h increased HO-1 expression and activity without affecting adiponectin expression and secretion. cobaltiprotoporphyrin 58-62 heme oxygenase 1 Homo sapiens 95-99 25917210-8 2015 Further, geraniin induced the expression of heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1) and level of glutathione (GSH) in a concentration- and time-dependent manner, and increased Nrf2 nuclear translocation. Geraniin 9-17 heme oxygenase 1 Homo sapiens 44-60 26405158-0 2015 Heme oxygenase-1 accelerates erastin-induced ferroptotic cell death. erastin 29-36 heme oxygenase 1 Homo sapiens 0-16 26405158-6 2015 Zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor, prevented Erastin-triggered ferroptotic cancer cell death. zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 33-37 26405158-6 2015 Zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor, prevented Erastin-triggered ferroptotic cancer cell death. zinc protoporphyrin 24-28 heme oxygenase 1 Homo sapiens 33-37 26405158-6 2015 Zinc protoporphyrin IX (ZnPP), a HO-1 inhibitor, prevented Erastin-triggered ferroptotic cancer cell death. erastin 59-66 heme oxygenase 1 Homo sapiens 33-37 26405158-7 2015 Furthermore, Erastin induced the protein and mRNA levels of HO-1 in HT-1080 fibrosarcoma cells. erastin 13-20 heme oxygenase 1 Homo sapiens 60-64 26405158-10 2015 In addition, hemin and CORM accelerate the HO-1 expression in the presence of Erastin and increase membranous lipid peroxidation. erastin 78-85 heme oxygenase 1 Homo sapiens 43-47 26405158-11 2015 Thus, HO-1 is an essential enzyme for iron-dependent lipid peroxidation during ferroptotic cell death. Iron 38-42 heme oxygenase 1 Homo sapiens 6-10 26366056-8 2015 Finally, we showed curcumin A induced expression of HO-1 and decreased cell cycle progression of T cells. curcumin a 19-29 heme oxygenase 1 Homo sapiens 52-56 25917210-8 2015 Further, geraniin induced the expression of heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1) and level of glutathione (GSH) in a concentration- and time-dependent manner, and increased Nrf2 nuclear translocation. Geraniin 9-17 heme oxygenase 1 Homo sapiens 62-66 25917210-10 2015 However, inhibitors of PI3K/AKT and ERK1/2 (LY294002 or U0126) not only suppressed geraniin-induced nuclear translocation of Nrf2 but also abolished the expression of HO-1, NQO1 and GSH. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 44-52 heme oxygenase 1 Homo sapiens 167-171 25917210-10 2015 However, inhibitors of PI3K/AKT and ERK1/2 (LY294002 or U0126) not only suppressed geraniin-induced nuclear translocation of Nrf2 but also abolished the expression of HO-1, NQO1 and GSH. U 0126 56-61 heme oxygenase 1 Homo sapiens 167-171 25917210-10 2015 However, inhibitors of PI3K/AKT and ERK1/2 (LY294002 or U0126) not only suppressed geraniin-induced nuclear translocation of Nrf2 but also abolished the expression of HO-1, NQO1 and GSH. Geraniin 83-91 heme oxygenase 1 Homo sapiens 167-171 25917210-11 2015 CONCLUSIONS: These results demonstrated that geraniin induced Nrf2-mediated expression of antioxidant enzymes HO-1 and NQO1, presumably via PI3K/AKT and ERK1/2 signaling pathways, thereby protecting cells from H2O2-induced oxidative cell death. Geraniin 45-53 heme oxygenase 1 Homo sapiens 110-114 25917210-11 2015 CONCLUSIONS: These results demonstrated that geraniin induced Nrf2-mediated expression of antioxidant enzymes HO-1 and NQO1, presumably via PI3K/AKT and ERK1/2 signaling pathways, thereby protecting cells from H2O2-induced oxidative cell death. Hydrogen Peroxide 210-214 heme oxygenase 1 Homo sapiens 110-114 25977183-0 2015 Z-ligustilide ameliorated ultraviolet B-induced oxidative stress and inflammatory cytokine production in human keratinocytes through upregulation of Nrf2/HO-1 and suppression of NF-kappaB pathway. ligustilide 0-13 heme oxygenase 1 Homo sapiens 154-158 26173399-4 2015 The inducible HO isoform (HO-1) has two functional genetic polymorphisms: (GT)n dinucleotide repeats and A(-413)T SNP (rs2071746), both of which can affect its promoter activity. Dinucleoside Phosphates 80-92 heme oxygenase 1 Homo sapiens 26-30 26617752-13 2015 ATRA decreased anti-oxygen protein Nrf2 and HO-1, and increases inflammatory factors NF-kappaB and TNF-alpha. Tretinoin 0-4 heme oxygenase 1 Homo sapiens 44-48 25977183-5 2015 Both silencing the nuclear factor E2-related factor 2 (Nrf2) and the supplement of tin protoporphyrin IX (SnPP), a haeme oxygenase-1 (HO-1) inhibitor, cancelled the inhibitory effect of Z-lig on UVB-induced ROS upregulation in NHEKs. tin protoporphyrin IX 83-104 heme oxygenase 1 Homo sapiens 115-139 25977183-5 2015 Both silencing the nuclear factor E2-related factor 2 (Nrf2) and the supplement of tin protoporphyrin IX (SnPP), a haeme oxygenase-1 (HO-1) inhibitor, cancelled the inhibitory effect of Z-lig on UVB-induced ROS upregulation in NHEKs. S-Nitroso-N-propionyl-D,L-penicillamine 106-110 heme oxygenase 1 Homo sapiens 115-139 25977183-5 2015 Both silencing the nuclear factor E2-related factor 2 (Nrf2) and the supplement of tin protoporphyrin IX (SnPP), a haeme oxygenase-1 (HO-1) inhibitor, cancelled the inhibitory effect of Z-lig on UVB-induced ROS upregulation in NHEKs. ligustilide 186-191 heme oxygenase 1 Homo sapiens 115-139 25977183-8 2015 Taken together, these findings suggest that Z-lig can suppress UVB-induced ROS generation through Nrf2/HO-1 upregulation and inflammation by suppressing the NF-kappaB pathway, suggesting that Z-lig may be beneficial in protecting skin from UVB exposure. ligustilide 44-49 heme oxygenase 1 Homo sapiens 103-107 25977183-8 2015 Taken together, these findings suggest that Z-lig can suppress UVB-induced ROS generation through Nrf2/HO-1 upregulation and inflammation by suppressing the NF-kappaB pathway, suggesting that Z-lig may be beneficial in protecting skin from UVB exposure. Reactive Oxygen Species 75-78 heme oxygenase 1 Homo sapiens 103-107 25977183-8 2015 Taken together, these findings suggest that Z-lig can suppress UVB-induced ROS generation through Nrf2/HO-1 upregulation and inflammation by suppressing the NF-kappaB pathway, suggesting that Z-lig may be beneficial in protecting skin from UVB exposure. ligustilide 192-197 heme oxygenase 1 Homo sapiens 103-107 26622665-11 2015 In addition, these results indicated that TET-dependent demethylation of the Nrf2 promoter upregulated Nrf2 and HO-1 expression, which induced cellular protection mechanisms, ultimately leading to drug resistance. tetramethylenedisulfotetramine 42-45 heme oxygenase 1 Homo sapiens 112-116 26550306-11 2015 CONCLUSION: Sevoflurane preconditioning can reduce oxidative stress injury induced by OLV and protect lung tissue by increasing HO-1 expression in lung tissue. Sevoflurane 12-23 heme oxygenase 1 Homo sapiens 128-132 26201059-7 2015 In the cardiovascular system, aspirin also acetylates eNOS with subsequent upregulation of NO formation and enhanced expression of the antioxidans heme-oxygenase-1. Aspirin 30-37 heme oxygenase 1 Homo sapiens 147-163 25922869-1 2015 BACKGROUND: In vitro and animal studies indicate that statins increase heme oxygenase-1 gene (HMOX1) expression, which then, presumably, increases plasma bilirubin concentration. Bilirubin 154-163 heme oxygenase 1 Homo sapiens 94-99 25922869-3 2015 We hypothesized that in patients with stable angina atorvastatin therapy (20 mg/day for 10 weeks) concomitantly increases total bilirubin concentration and HMOX1 expression, as assessed non-invasively by plasma analysis. Atorvastatin 52-64 heme oxygenase 1 Homo sapiens 156-161 26071936-4 2015 We previously demonstrated that curcumin increased reactive oxygen species (ROS) formation and apoptosis in dermal fibroblasts, which could be prevented by pre-induction of the cytoprotective enzyme heme oxygenase (HO)-1. Curcumin 32-40 heme oxygenase 1 Homo sapiens 199-220 26071936-4 2015 We previously demonstrated that curcumin increased reactive oxygen species (ROS) formation and apoptosis in dermal fibroblasts, which could be prevented by pre-induction of the cytoprotective enzyme heme oxygenase (HO)-1. Reactive Oxygen Species 51-74 heme oxygenase 1 Homo sapiens 199-220 26071936-4 2015 We previously demonstrated that curcumin increased reactive oxygen species (ROS) formation and apoptosis in dermal fibroblasts, which could be prevented by pre-induction of the cytoprotective enzyme heme oxygenase (HO)-1. Reactive Oxygen Species 76-79 heme oxygenase 1 Homo sapiens 199-220 25994789-6 2015 Active Nrf2 and the antioxidant enzyme heme oxygenase-1 (HO-1) increase at 3 h after tBHQ treatment. 2-tert-butylhydroquinone 85-89 heme oxygenase 1 Homo sapiens 39-55 25998388-5 2015 Since the mode of action of hemin is both physiological and pharmacological through induction of heme oxygenase-1 (HO-1), an endogenous host protective response to an FDA-licensed therapeutic used to treat another disease, our study suggests an approach to developing novel, safe and effective therapeutic strategies for treating bone disorders, because hemin administration in humans has previously met required FDA safety standards. Hemin 28-33 heme oxygenase 1 Homo sapiens 97-113 25998388-5 2015 Since the mode of action of hemin is both physiological and pharmacological through induction of heme oxygenase-1 (HO-1), an endogenous host protective response to an FDA-licensed therapeutic used to treat another disease, our study suggests an approach to developing novel, safe and effective therapeutic strategies for treating bone disorders, because hemin administration in humans has previously met required FDA safety standards. Hemin 28-33 heme oxygenase 1 Homo sapiens 115-119 25998388-5 2015 Since the mode of action of hemin is both physiological and pharmacological through induction of heme oxygenase-1 (HO-1), an endogenous host protective response to an FDA-licensed therapeutic used to treat another disease, our study suggests an approach to developing novel, safe and effective therapeutic strategies for treating bone disorders, because hemin administration in humans has previously met required FDA safety standards. Hemin 354-359 heme oxygenase 1 Homo sapiens 97-113 25998388-5 2015 Since the mode of action of hemin is both physiological and pharmacological through induction of heme oxygenase-1 (HO-1), an endogenous host protective response to an FDA-licensed therapeutic used to treat another disease, our study suggests an approach to developing novel, safe and effective therapeutic strategies for treating bone disorders, because hemin administration in humans has previously met required FDA safety standards. Hemin 354-359 heme oxygenase 1 Homo sapiens 115-119 26309414-0 2015 Heme oxygenase-1 is a predictive biomarker for therapeutic targeting of advanced clear cell renal cell carcinoma treated with sorafenib or sunitinib. Sorafenib 126-135 heme oxygenase 1 Homo sapiens 0-16 26309414-0 2015 Heme oxygenase-1 is a predictive biomarker for therapeutic targeting of advanced clear cell renal cell carcinoma treated with sorafenib or sunitinib. Sunitinib 139-148 heme oxygenase 1 Homo sapiens 0-16 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sorafenib 228-237 heme oxygenase 1 Homo sapiens 42-58 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sorafenib 228-237 heme oxygenase 1 Homo sapiens 60-64 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sunitinib 242-251 heme oxygenase 1 Homo sapiens 42-58 26309414-1 2015 BACKGROUND: We analyzed the expression of heme oxygenase-1 (HO-1) in patients undergoing radical nephrectomy for advanced clear cell renal cell carcinoma (CC-RCC) and evaluated the effects of the targeted therapies treated with sorafenib and sunitinib. Sunitinib 242-251 heme oxygenase 1 Homo sapiens 60-64 26309414-6 2015 In the low HO-1 level group, a higher tumor response rate and a longer survival time was achieved in patients who received sorafenib or sunitinib. Sorafenib 123-132 heme oxygenase 1 Homo sapiens 11-15 26309414-6 2015 In the low HO-1 level group, a higher tumor response rate and a longer survival time was achieved in patients who received sorafenib or sunitinib. Sunitinib 136-145 heme oxygenase 1 Homo sapiens 11-15 26309414-9 2015 Overall, HO-1 expression might be a useful biomarker for predicting the response to sunitinib and sorafenib for patients with metastatic CC-RCC. Sunitinib 84-93 heme oxygenase 1 Homo sapiens 9-13 26309414-9 2015 Overall, HO-1 expression might be a useful biomarker for predicting the response to sunitinib and sorafenib for patients with metastatic CC-RCC. Sorafenib 98-107 heme oxygenase 1 Homo sapiens 9-13 26163453-0 2015 Insight into the molecular mechanism of heme oxygenase-1 induction by docosahexaenoic acid in U937 cells. Docosahexaenoic Acids 70-90 heme oxygenase 1 Homo sapiens 40-56 26163453-2 2015 To date, little is known about whether fatty acids can affect HO-1 induction. Fatty Acids 39-50 heme oxygenase 1 Homo sapiens 62-66 26163453-3 2015 Here, we report the induction of HO-1 by docosahexaenoic acid (DHA) and the associated molecular mechanisms in human myelomonocytic lymphoma U937 cells. Docosahexaenoic Acids 41-61 heme oxygenase 1 Homo sapiens 33-37 26163453-3 2015 Here, we report the induction of HO-1 by docosahexaenoic acid (DHA) and the associated molecular mechanisms in human myelomonocytic lymphoma U937 cells. Docosahexaenoic Acids 63-66 heme oxygenase 1 Homo sapiens 33-37 26163453-4 2015 When U937 cells were treated with DHA, eicosapentaenoic acid, palmitic acid or oleic acid, DHA was the most effective inducer of HO-1. Docosahexaenoic Acids 34-37 heme oxygenase 1 Homo sapiens 129-133 26163453-4 2015 When U937 cells were treated with DHA, eicosapentaenoic acid, palmitic acid or oleic acid, DHA was the most effective inducer of HO-1. Eicosapentaenoic Acid 39-60 heme oxygenase 1 Homo sapiens 129-133 26163453-4 2015 When U937 cells were treated with DHA, eicosapentaenoic acid, palmitic acid or oleic acid, DHA was the most effective inducer of HO-1. Palmitic Acid 62-75 heme oxygenase 1 Homo sapiens 129-133 26163453-4 2015 When U937 cells were treated with DHA, eicosapentaenoic acid, palmitic acid or oleic acid, DHA was the most effective inducer of HO-1. Oleic Acid 79-89 heme oxygenase 1 Homo sapiens 129-133 26163453-4 2015 When U937 cells were treated with DHA, eicosapentaenoic acid, palmitic acid or oleic acid, DHA was the most effective inducer of HO-1. Docosahexaenoic Acids 91-94 heme oxygenase 1 Homo sapiens 129-133 26163453-7 2015 The increase in HO-1 expression was significantly inhibited by U0126, an ERK1/2 inhibitor. U 0126 63-68 heme oxygenase 1 Homo sapiens 16-20 26163453-8 2015 Nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf-2) and its binding to the HO-1 promoter significantly increased upon DHA treatment. Docosahexaenoic Acids 143-146 heme oxygenase 1 Homo sapiens 100-104 26163453-10 2015 Pretreatment with NAC dramatically inhibited the ERK1/2 activation, binding of Nrf-2 to antioxidant response elements (AREs) located in the HO-1 promoter and the induction of HO-1 by DHA. Docosahexaenoic Acids 183-186 heme oxygenase 1 Homo sapiens 175-179 26163453-11 2015 In conclusion, DHA increased HO-1 expression in U937 cells via activation of ERK1/2 and increased Nrf-2 binding to ARE in the HO-1 promoter. Docosahexaenoic Acids 15-18 heme oxygenase 1 Homo sapiens 29-33 26163453-11 2015 In conclusion, DHA increased HO-1 expression in U937 cells via activation of ERK1/2 and increased Nrf-2 binding to ARE in the HO-1 promoter. Docosahexaenoic Acids 15-18 heme oxygenase 1 Homo sapiens 126-130 25994789-7 2015 Chelating intracellular free zinc with tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) during tBHQ exposure partially abrogates the tBHQ-induced activation of Nrf2 and HO-1 expression, while Keap1 is further decreased. N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine 82-86 heme oxygenase 1 Homo sapiens 169-173 25994789-7 2015 Chelating intracellular free zinc with tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) during tBHQ exposure partially abrogates the tBHQ-induced activation of Nrf2 and HO-1 expression, while Keap1 is further decreased. 2-tert-butylhydroquinone 133-137 heme oxygenase 1 Homo sapiens 169-173 26270345-1 2015 Earlier observations indicate that free heme is selectively toxic to cells lacking heme oxygenase-1 (HO-1) but how this enzyme prevents heme toxicity remains unexplained. Heme 40-44 heme oxygenase 1 Homo sapiens 83-99 26270345-1 2015 Earlier observations indicate that free heme is selectively toxic to cells lacking heme oxygenase-1 (HO-1) but how this enzyme prevents heme toxicity remains unexplained. Heme 40-44 heme oxygenase 1 Homo sapiens 101-105 26270345-2 2015 Here, using A549 (human lung cancer) and immortalized human bronchial epithelial cells incubated with exogenous heme, we find knock-down of HO-1 using siRNA does promote the accumulation of cell-associated heme and heme-induced cell death. Heme 112-116 heme oxygenase 1 Homo sapiens 140-144 26270345-2 2015 Here, using A549 (human lung cancer) and immortalized human bronchial epithelial cells incubated with exogenous heme, we find knock-down of HO-1 using siRNA does promote the accumulation of cell-associated heme and heme-induced cell death. Heme 206-210 heme oxygenase 1 Homo sapiens 140-144 26270345-2 2015 Here, using A549 (human lung cancer) and immortalized human bronchial epithelial cells incubated with exogenous heme, we find knock-down of HO-1 using siRNA does promote the accumulation of cell-associated heme and heme-induced cell death. Heme 206-210 heme oxygenase 1 Homo sapiens 140-144 26270345-3 2015 However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Heme 46-50 heme oxygenase 1 Homo sapiens 173-177 26270345-3 2015 However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Iron 100-104 heme oxygenase 1 Homo sapiens 173-177 26270345-3 2015 However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Heme 134-138 heme oxygenase 1 Homo sapiens 173-177 26270345-3 2015 However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Deferoxamine 199-214 heme oxygenase 1 Homo sapiens 173-177 26270345-5 2015 Supporting the importance of endogenous oxidant production, both chemical and siRNA inhibition of catalase activity predisposes HO-1 deficient cells to heme-mediated killing. Heme 152-156 heme oxygenase 1 Homo sapiens 128-132 26270345-6 2015 Importantly, it appears that HO-1 deficiency somehow blocks the induction of ferritin; control cells exposed to heme show ~10-fold increases in ferritin heavy chain expression whereas in heme-exposed HO-1 deficient cells ferritin expression is unchanged. Heme 112-116 heme oxygenase 1 Homo sapiens 29-33 26270345-6 2015 Importantly, it appears that HO-1 deficiency somehow blocks the induction of ferritin; control cells exposed to heme show ~10-fold increases in ferritin heavy chain expression whereas in heme-exposed HO-1 deficient cells ferritin expression is unchanged. Heme 187-191 heme oxygenase 1 Homo sapiens 29-33 26270345-7 2015 Finally, overexpression of ferritin H chain in HO-1 deficient cells completely prevents heme-induced cytotoxicity. Heme 88-92 heme oxygenase 1 Homo sapiens 47-51 26270345-8 2015 Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 31-35 26270345-8 2015 Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 93-97 26270345-8 2015 Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 93-97 26270345-8 2015 Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 93-97 26208625-0 2015 How does binding of imidazole-based inhibitors to heme oxygenase-1 influence their conformation? imidazole 20-29 heme oxygenase 1 Homo sapiens 50-66 26208625-3 2015 Imidazole-based analogues show effective and selective inhibitory potency of HO-1. imidazole 0-9 heme oxygenase 1 Homo sapiens 77-81 26208625-6 2015 Molecular docking of imidazole-based analogues in the active site of HO-1 is in good agreement with the experimental structures. imidazole 21-30 heme oxygenase 1 Homo sapiens 69-73 26208625-7 2015 Inhibitors interact with the heme cofactor and a hydrophobic pocket (Met34, Phe37, Val50, Leu147 and Phe214) in the HO-1 binding site. Heme 29-33 heme oxygenase 1 Homo sapiens 116-120 25994789-6 2015 Active Nrf2 and the antioxidant enzyme heme oxygenase-1 (HO-1) increase at 3 h after tBHQ treatment. 2-tert-butylhydroquinone 85-89 heme oxygenase 1 Homo sapiens 57-61 25994789-7 2015 Chelating intracellular free zinc with tetrakis-(2-pyridylmethyl)ethylenediamine (TPEN) during tBHQ exposure partially abrogates the tBHQ-induced activation of Nrf2 and HO-1 expression, while Keap1 is further decreased. SCHEMBL231091 39-80 heme oxygenase 1 Homo sapiens 169-173 26080289-7 2015 This resistance was due to an ability to cope with PENAO-induced oxidative stress, notably through heme oxygenase-1 induction, and a shift in metabolism towards glycolysis. HH7VUN8QBQ 51-56 heme oxygenase 1 Homo sapiens 99-115 26041409-0 2015 Src/STAT3-dependent heme oxygenase-1 induction mediates chemoresistance of breast cancer cells to doxorubicin by promoting autophagy. Doxorubicin 98-109 heme oxygenase 1 Homo sapiens 20-36 26041409-3 2015 In the present study, we found an induction of HO-1 expression in doxorubicin (DOX)-treated MDA-MB-231 human breast adenocarcinoma cells, which showed insensitivity to DOX treatment. Doxorubicin 66-77 heme oxygenase 1 Homo sapiens 47-51 26041409-3 2015 In the present study, we found an induction of HO-1 expression in doxorubicin (DOX)-treated MDA-MB-231 human breast adenocarcinoma cells, which showed insensitivity to DOX treatment. Doxorubicin 79-82 heme oxygenase 1 Homo sapiens 47-51 26041409-4 2015 Knockdown HO-1 expression dramatically upregulated the incidence of MDA-MB-231 cell death under DOX treatment, indicating that HO-1 functions as a critical contributor to drug resistance in MDA-MB-231 cells. Doxorubicin 96-99 heme oxygenase 1 Homo sapiens 10-14 26041409-4 2015 Knockdown HO-1 expression dramatically upregulated the incidence of MDA-MB-231 cell death under DOX treatment, indicating that HO-1 functions as a critical contributor to drug resistance in MDA-MB-231 cells. Doxorubicin 96-99 heme oxygenase 1 Homo sapiens 127-131 26041409-5 2015 We further observed that DOX exposure induced a cytoprotective autophagic flux in MDA-MB-231 cells, which was dependent on HO-1 induction. Doxorubicin 25-28 heme oxygenase 1 Homo sapiens 123-127 26041409-8 2015 Therefore, we conclude that Src/STAT3-dependent HO-1 induction protects MDA-MB-231 breast cancer cells from DOX-induced death through promoting autophagy. Doxorubicin 108-111 heme oxygenase 1 Homo sapiens 48-52 26041409-9 2015 In the following study, we further demonstrated the contribution of Src/STAT3/HO-1/autophagy pathway activation to DOX resistance in another breast cancer cell line, MDA-MB-468, which bears a similar phenotype to MDA-MB-231 cells. Doxorubicin 115-118 heme oxygenase 1 Homo sapiens 78-82 26041409-10 2015 Therefore, activation of Src/STAT3/HO-1/autophagy signaling pathway might play a general role in protecting certain subtypes of breast cancer cells from DOX-induced cytotoxicity. Doxorubicin 153-156 heme oxygenase 1 Homo sapiens 35-39 25881548-4 2015 Quercetin induced the nuclear translocation of Nrf2, along with the increased expression of the antioxidant responsive element (ARE)-dependent genes like catalytic or modify subunit of glutamate-cysteine ligase (GCLC/GCLM), and heme oxygenase-1 (HO-1). Quercetin 0-9 heme oxygenase 1 Homo sapiens 228-244 25881548-4 2015 Quercetin induced the nuclear translocation of Nrf2, along with the increased expression of the antioxidant responsive element (ARE)-dependent genes like catalytic or modify subunit of glutamate-cysteine ligase (GCLC/GCLM), and heme oxygenase-1 (HO-1). Quercetin 0-9 heme oxygenase 1 Homo sapiens 246-250 25881548-5 2015 In addition, the HO-1 inhibitor zinc protoporphyrin (ZnPP) and the GCL inhibitor L-buthionine-(S,R)-sulfoximine (BSO) both reduced the hepatoprotection induced by quercetin. zinc protoporphyrin 32-51 heme oxygenase 1 Homo sapiens 17-21 25881548-5 2015 In addition, the HO-1 inhibitor zinc protoporphyrin (ZnPP) and the GCL inhibitor L-buthionine-(S,R)-sulfoximine (BSO) both reduced the hepatoprotection induced by quercetin. zinc protoporphyrin 53-57 heme oxygenase 1 Homo sapiens 17-21 25881548-5 2015 In addition, the HO-1 inhibitor zinc protoporphyrin (ZnPP) and the GCL inhibitor L-buthionine-(S,R)-sulfoximine (BSO) both reduced the hepatoprotection induced by quercetin. Quercetin 163-172 heme oxygenase 1 Homo sapiens 17-21 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 93-109 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 111-115 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 93-109 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 111-115 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. metallo-protoporphyrin 34-56 heme oxygenase 1 Homo sapiens 93-109 25981584-1 2015 Cobalt protoporphyrin (CoPP) is a metallo-protoporphyrin that works as a powerful inducer of heme oxigenase-1 (HO-1) in various tissues and cells. metallo-protoporphyrin 34-56 heme oxygenase 1 Homo sapiens 111-115 25881548-11 2015 In addition, SP600125 also decreased the increased mRNA and protein expression of GCLC, GCLM, and HO-1 induced by quercetin. pyrazolanthrone 13-21 heme oxygenase 1 Homo sapiens 98-102 25881548-11 2015 In addition, SP600125 also decreased the increased mRNA and protein expression of GCLC, GCLM, and HO-1 induced by quercetin. Quercetin 114-123 heme oxygenase 1 Homo sapiens 98-102 26241392-9 2015 Mechanistically, 4-AC blocked the increase of cellular ROS induced by oxidative stress, and upregulated NQO1 and HO-1 genes by stabilizing and inducing the nuclear translocation of NRF2 transcription factor. 4-hydroxyphenylacetate 17-21 heme oxygenase 1 Homo sapiens 113-117 26283888-0 2015 Arsenic Trioxide Activate Transcription of Heme Oxygenase-1 by Promoting Nuclear Translocation of NFE2L2. Arsenic Trioxide 0-16 heme oxygenase 1 Homo sapiens 43-59 26283888-1 2015 In a previous study, we found that induced expression of Heme Oxygenase-1 (HO-1) is responsible for the resistance of human osteosarcoma MG63 cells to the chemotherapeutic agent arsenic trioxide (ATO). Arsenic Trioxide 178-194 heme oxygenase 1 Homo sapiens 57-73 26283888-1 2015 In a previous study, we found that induced expression of Heme Oxygenase-1 (HO-1) is responsible for the resistance of human osteosarcoma MG63 cells to the chemotherapeutic agent arsenic trioxide (ATO). Arsenic Trioxide 178-194 heme oxygenase 1 Homo sapiens 75-79 26283888-1 2015 In a previous study, we found that induced expression of Heme Oxygenase-1 (HO-1) is responsible for the resistance of human osteosarcoma MG63 cells to the chemotherapeutic agent arsenic trioxide (ATO). Arsenic Trioxide 196-199 heme oxygenase 1 Homo sapiens 57-73 26283888-1 2015 In a previous study, we found that induced expression of Heme Oxygenase-1 (HO-1) is responsible for the resistance of human osteosarcoma MG63 cells to the chemotherapeutic agent arsenic trioxide (ATO). Arsenic Trioxide 196-199 heme oxygenase 1 Homo sapiens 75-79 26283888-2 2015 The present study was aimed at investigating the molecular mechanisms underlying the induction of HO-1 that occurs after exposure of MG63 cells to ATO. Arsenic Trioxide 147-150 heme oxygenase 1 Homo sapiens 98-102 26283888-3 2015 First, using RT-QPCT and Western-blot, we found that ATO strongly induced the expression of heme oxygenase-1 (HO-1) in these human osteosarcoma cells. Arsenic Trioxide 53-56 heme oxygenase 1 Homo sapiens 92-108 26283888-3 2015 First, using RT-QPCT and Western-blot, we found that ATO strongly induced the expression of heme oxygenase-1 (HO-1) in these human osteosarcoma cells. Arsenic Trioxide 53-56 heme oxygenase 1 Homo sapiens 110-114 26283888-4 2015 Then by analyzing HO-1 mRNA of MG63 cells exposed to ATO in the presence and absence of a transcription inhibitor Actinomycin-D (Act-D), we demonstrated that ATO activates HO-1 expression in MG63 cells by regulating the transcription of the gene. Arsenic Trioxide 53-56 heme oxygenase 1 Homo sapiens 18-22 26283888-4 2015 Then by analyzing HO-1 mRNA of MG63 cells exposed to ATO in the presence and absence of a transcription inhibitor Actinomycin-D (Act-D), we demonstrated that ATO activates HO-1 expression in MG63 cells by regulating the transcription of the gene. Arsenic Trioxide 53-56 heme oxygenase 1 Homo sapiens 172-176 26283888-4 2015 Then by analyzing HO-1 mRNA of MG63 cells exposed to ATO in the presence and absence of a transcription inhibitor Actinomycin-D (Act-D), we demonstrated that ATO activates HO-1 expression in MG63 cells by regulating the transcription of the gene. Dactinomycin 114-127 heme oxygenase 1 Homo sapiens 172-176 26283888-4 2015 Then by analyzing HO-1 mRNA of MG63 cells exposed to ATO in the presence and absence of a transcription inhibitor Actinomycin-D (Act-D), we demonstrated that ATO activates HO-1 expression in MG63 cells by regulating the transcription of the gene. Arsenic Trioxide 158-161 heme oxygenase 1 Homo sapiens 18-22 26283888-4 2015 Then by analyzing HO-1 mRNA of MG63 cells exposed to ATO in the presence and absence of a transcription inhibitor Actinomycin-D (Act-D), we demonstrated that ATO activates HO-1 expression in MG63 cells by regulating the transcription of the gene. Arsenic Trioxide 158-161 heme oxygenase 1 Homo sapiens 172-176 26283888-6 2015 From these results we have concluded that transcription activation of HO-1 resulting from the nuclear translocation of NFE2L2 is the underlying molecular mechanism for its high induction, which, in turn, is responsible for the resistance of human osteosarcoma cells to ATO treatment. Arsenic Trioxide 269-272 heme oxygenase 1 Homo sapiens 70-74 26082074-2 2015 In the present study, in order to elucidate the mechanisms responsible for the antioxidant activity of GPE, its effects on the expression of critical antioxidant enzymes, such as catalase (CAT), superoxide dismutase (SOD)1, heme oxygenase 1 (HO-1) and gamma-glutamylcysteine synthetase (GCS) were assessed in EA.hy926 and C2C12 cells. gpe 103-106 heme oxygenase 1 Homo sapiens 224-240 26267493-0 2015 Effect of heat stress-induced production of mitochondrial reactive oxygen species on NADPH oxidase and heme oxygenase-1 mRNA levels in avian muscle cells. Reactive Oxygen Species 58-81 heme oxygenase 1 Homo sapiens 103-119 26267493-6 2015 Addition of the superoxide scavenger 4-hydroxy TEMPO to the culture medium of heat-stressed cells restored mitochondrial superoxide and intracellular ROS levels as well as NOX4 and HO-1 mRNA levels to near-normal values. Superoxides 16-26 heme oxygenase 1 Homo sapiens 181-185 26267493-6 2015 Addition of the superoxide scavenger 4-hydroxy TEMPO to the culture medium of heat-stressed cells restored mitochondrial superoxide and intracellular ROS levels as well as NOX4 and HO-1 mRNA levels to near-normal values. TEMPOL-H 37-52 heme oxygenase 1 Homo sapiens 181-185 26267493-7 2015 We suggest that mitochondrial superoxide production could play an influential role in augmenting oxidative damage to avian muscle cells, possibly via the up-regulation of NOX4 and down-regulation of HO-1 in heat-stressed avian muscle cells. Superoxides 30-40 heme oxygenase 1 Homo sapiens 199-203 25951827-9 2015 Genetic silencing of Nrf2 prevented the induction of HO-1 and mitochondrial biogenesis by cilostazol in HepG2 cells. Cilostazol 90-100 heme oxygenase 1 Homo sapiens 53-57 26211586-0 2015 Cerium oxide nanoparticles alleviate oxidative stress and decreases Nrf-2/HO-1 in D-GALN/LPS induced hepatotoxicity. ceric oxide 0-12 heme oxygenase 1 Homo sapiens 74-78 26211586-1 2015 Translocation of the master regulator of antioxidant-response element-driven antioxidant gene, nuclear factor erythroid 2 (Nrf-2) from the cytoplasm into the nucleus and triggering the transcription of hemoxygenase-1 (HO-1) to counteract the oxidative stress is a key feature in D-galactoseamine and lipopolysaccharide (D-GALN/LPS) induced hepatotoxicity. d-galactoseamine 279-295 heme oxygenase 1 Homo sapiens 202-222 26088684-0 2015 Resveratrol Protects Hippocampal Astrocytes Against LPS-Induced Neurotoxicity Through HO-1, p38 and ERK Pathways. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 86-90 26088684-11 2015 Additionally, we demonstrated the involvement of NFkappaB, HO-1, p38 and ERK signaling pathways in the protective effect of resveratrol, providing the first mechanistic explanation for these effects in hippocampal astrocytes. Resveratrol 124-135 heme oxygenase 1 Homo sapiens 59-63 26146841-3 2015 Eleven common mutations and polymorphisms across five bilirubin metabolism genes, namely those encoding UGT1A1, HMOX1, BLVRA, SLCO1B1 and SLCO1B3, were determined using the high resolution melt (HRM) assay or PCR-capillary electrophoresis analysis. Bilirubin 54-63 heme oxygenase 1 Homo sapiens 112-117 26211586-2 2015 We mainly aimed to study the effect of cerium oxide (CeO2) nanoparticles on Nrf-2/HO-1 pathway whereas; it has previously shown to have an antioxidant effect in liver models. ceric oxide 39-51 heme oxygenase 1 Homo sapiens 82-86 26211586-2 2015 We mainly aimed to study the effect of cerium oxide (CeO2) nanoparticles on Nrf-2/HO-1 pathway whereas; it has previously shown to have an antioxidant effect in liver models. ceric oxide 53-57 heme oxygenase 1 Homo sapiens 82-86 26211586-3 2015 Administration of CeO2 nanoparticles significantly decreased the translocation of the cytoplasmic Nrf-2 with a concomitant decrement in the gene expression of HO-1 as it reveals a powerful antioxidative effect as indicated by the significant increase in the levels of glutathione (GSH), glutathione peroxidase (GPX1), glutathione reductase (GR), superoxide dismutase (SOD) and catalase. Glutathione 268-279 heme oxygenase 1 Homo sapiens 159-163 26211586-3 2015 Administration of CeO2 nanoparticles significantly decreased the translocation of the cytoplasmic Nrf-2 with a concomitant decrement in the gene expression of HO-1 as it reveals a powerful antioxidative effect as indicated by the significant increase in the levels of glutathione (GSH), glutathione peroxidase (GPX1), glutathione reductase (GR), superoxide dismutase (SOD) and catalase. Glutathione 281-284 heme oxygenase 1 Homo sapiens 159-163 25109682-0 2015 Monosodium urate crystals trigger Nrf2- and heme oxygenase-1-dependent inflammation in THP-1 cells. Uric Acid 0-16 heme oxygenase 1 Homo sapiens 44-60 25109682-9 2015 MSU-induced IL-1beta secretion and NLRP3 inflammasome activation were inhibited by the knockdown of Nrf2 and via the HO-1 inhibitor zinc (II) protoporphyrin IX (ZnPP). zinc;3-[7,12-bis(ethenyl)-18-(3-hydroperoxyprop-1-en-2-yl)-3,8,13,17-tetramethylporphyrin-21,23-diid-2-yl]propanoic acid 132-159 heme oxygenase 1 Homo sapiens 117-121 25109682-9 2015 MSU-induced IL-1beta secretion and NLRP3 inflammasome activation were inhibited by the knockdown of Nrf2 and via the HO-1 inhibitor zinc (II) protoporphyrin IX (ZnPP). zinc protoporphyrin 161-165 heme oxygenase 1 Homo sapiens 117-121 25109682-10 2015 In addition, HO-1 inhibition increased the level of superoxide anion production and the consumption of glutathione. Superoxides 52-68 heme oxygenase 1 Homo sapiens 13-17 25109682-10 2015 In addition, HO-1 inhibition increased the level of superoxide anion production and the consumption of glutathione. Glutathione 103-114 heme oxygenase 1 Homo sapiens 13-17 25109682-11 2015 These findings suggest that Nrf2 and HO-1 mediate redox homeostasis and interact with pro-inflammatory factors in MSU-challenged THP-1 cells, thereby providing new insight into how MSU-induced gouty inflammation is mediated by specific mechanisms that are involved in the Nrf2/Ho-1 antioxidant signaling pathway.Cellular & Molecular Immunology advance online publication, 11 August 2014; doi:10.1038/cmi.2014.65. Uric Acid 114-117 heme oxygenase 1 Homo sapiens 37-41 25109682-11 2015 These findings suggest that Nrf2 and HO-1 mediate redox homeostasis and interact with pro-inflammatory factors in MSU-challenged THP-1 cells, thereby providing new insight into how MSU-induced gouty inflammation is mediated by specific mechanisms that are involved in the Nrf2/Ho-1 antioxidant signaling pathway.Cellular & Molecular Immunology advance online publication, 11 August 2014; doi:10.1038/cmi.2014.65. Uric Acid 114-117 heme oxygenase 1 Homo sapiens 277-281 25109682-11 2015 These findings suggest that Nrf2 and HO-1 mediate redox homeostasis and interact with pro-inflammatory factors in MSU-challenged THP-1 cells, thereby providing new insight into how MSU-induced gouty inflammation is mediated by specific mechanisms that are involved in the Nrf2/Ho-1 antioxidant signaling pathway.Cellular & Molecular Immunology advance online publication, 11 August 2014; doi:10.1038/cmi.2014.65. Adenosine Monophosphate 322-325 heme oxygenase 1 Homo sapiens 37-41 25820186-1 2015 BACKGROUND: Over-expression of the heme-degrading enzyme, heme oxygenase-1 (HO-1) promotes iron deposition, mitochondrial damage, and autophagy in astrocytes and enhances the vulnerability of nearby neuronal constituents to oxidative injury. Heme 35-39 heme oxygenase 1 Homo sapiens 58-74 25820186-1 2015 BACKGROUND: Over-expression of the heme-degrading enzyme, heme oxygenase-1 (HO-1) promotes iron deposition, mitochondrial damage, and autophagy in astrocytes and enhances the vulnerability of nearby neuronal constituents to oxidative injury. Heme 35-39 heme oxygenase 1 Homo sapiens 76-80 25820186-1 2015 BACKGROUND: Over-expression of the heme-degrading enzyme, heme oxygenase-1 (HO-1) promotes iron deposition, mitochondrial damage, and autophagy in astrocytes and enhances the vulnerability of nearby neuronal constituents to oxidative injury. Iron 91-95 heme oxygenase 1 Homo sapiens 58-74 25820186-1 2015 BACKGROUND: Over-expression of the heme-degrading enzyme, heme oxygenase-1 (HO-1) promotes iron deposition, mitochondrial damage, and autophagy in astrocytes and enhances the vulnerability of nearby neuronal constituents to oxidative injury. Iron 91-95 heme oxygenase 1 Homo sapiens 76-80 25820186-7 2015 CO and Fe were implicated in the HMOX1 effects, whereas bilirubin was inert or counteracted the HMOX1-related changes. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 33-38 25820186-7 2015 CO and Fe were implicated in the HMOX1 effects, whereas bilirubin was inert or counteracted the HMOX1-related changes. Bilirubin 56-65 heme oxygenase 1 Homo sapiens 96-101 26011640-8 2015 Collectively, we found that microglial HO-1 and the generation of CO are essential for effective elimination of blood and heme after SAH that otherwise leads to neuronal injury and cognitive dysfunction. Heme 122-126 heme oxygenase 1 Homo sapiens 39-43 25929446-7 2015 In addition, phloretin increased heme oxygenase-1 production in a concentration-dependent manner. Phloretin 13-22 heme oxygenase 1 Homo sapiens 33-49 26595496-2 2015 Heme oxygenase-1(HO-1) metabolizes heme into biliverdin, bilirubin, carbon monoxide, and iron, our recent study showed that serum level of HO-1 was increased in stroke patients, yet the association of HO-1 level with risk of intracerebral hemorrhage (ICH) is poorly known. Heme 35-39 heme oxygenase 1 Homo sapiens 0-16 26595496-2 2015 Heme oxygenase-1(HO-1) metabolizes heme into biliverdin, bilirubin, carbon monoxide, and iron, our recent study showed that serum level of HO-1 was increased in stroke patients, yet the association of HO-1 level with risk of intracerebral hemorrhage (ICH) is poorly known. Biliverdine 45-55 heme oxygenase 1 Homo sapiens 0-16 26595496-2 2015 Heme oxygenase-1(HO-1) metabolizes heme into biliverdin, bilirubin, carbon monoxide, and iron, our recent study showed that serum level of HO-1 was increased in stroke patients, yet the association of HO-1 level with risk of intracerebral hemorrhage (ICH) is poorly known. Bilirubin 57-66 heme oxygenase 1 Homo sapiens 0-16 26595496-2 2015 Heme oxygenase-1(HO-1) metabolizes heme into biliverdin, bilirubin, carbon monoxide, and iron, our recent study showed that serum level of HO-1 was increased in stroke patients, yet the association of HO-1 level with risk of intracerebral hemorrhage (ICH) is poorly known. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 0-16 26595496-2 2015 Heme oxygenase-1(HO-1) metabolizes heme into biliverdin, bilirubin, carbon monoxide, and iron, our recent study showed that serum level of HO-1 was increased in stroke patients, yet the association of HO-1 level with risk of intracerebral hemorrhage (ICH) is poorly known. Iron 89-93 heme oxygenase 1 Homo sapiens 0-16 25815672-0 2015 Effects of Eicosapentaenoic Acid on the Cytoprotection Through Nrf2-Mediated Heme Oxygenase-1 in Human Endothelial Cells. Eicosapentaenoic Acid 11-32 heme oxygenase 1 Homo sapiens 77-93 25815672-2 2015 The aim of this study was to investigate whether heme oxygenase-1 (HO-1) induction contributes to the cytoprotective effects of EPA in endothelial cells threatened with oxidative damage. Eicosapentaenoic Acid 128-131 heme oxygenase 1 Homo sapiens 49-65 25815672-2 2015 The aim of this study was to investigate whether heme oxygenase-1 (HO-1) induction contributes to the cytoprotective effects of EPA in endothelial cells threatened with oxidative damage. Eicosapentaenoic Acid 128-131 heme oxygenase 1 Homo sapiens 67-71 25804888-10 2015 Increased transcript expression of metallothioneins, MT1A and MT2A and the stress response genes HMOX1 (690%) and HIF1A (247%) occurred in CATLE cells possibly in adaptation to chronic arsenic exposure. Arsenic 185-192 heme oxygenase 1 Homo sapiens 97-102 25815672-3 2015 In this study, we investigated the effect of EPA on the induction of HO-1 by NF-E2-related factor 2 (Nrf2) in human umbilical vein endothelial cells. Eicosapentaenoic Acid 45-48 heme oxygenase 1 Homo sapiens 69-73 25815672-4 2015 In cells treated with low concentrations of EPA (10-25 muM), HO-1 expression increased in a time- and concentration-dependent manner. Eicosapentaenoic Acid 44-47 heme oxygenase 1 Homo sapiens 61-65 25815672-5 2015 Additionally, EPA treatment increased Nrf2 nuclear translocation and antioxidant response element activity, leading to the upregulation of HO-1 expression. Eicosapentaenoic Acid 14-17 heme oxygenase 1 Homo sapiens 139-143 25815672-7 2015 The reduction in cell death was reversed by treatment with zinc protoporphyrin, an inhibitor of HO-1, indicating that HO-1 contributed to the protective effect of EPA. zinc protoporphyrin 59-78 heme oxygenase 1 Homo sapiens 96-100 25815672-7 2015 The reduction in cell death was reversed by treatment with zinc protoporphyrin, an inhibitor of HO-1, indicating that HO-1 contributed to the protective effect of EPA. zinc protoporphyrin 59-78 heme oxygenase 1 Homo sapiens 118-122 25815672-7 2015 The reduction in cell death was reversed by treatment with zinc protoporphyrin, an inhibitor of HO-1, indicating that HO-1 contributed to the protective effect of EPA. Eicosapentaenoic Acid 163-166 heme oxygenase 1 Homo sapiens 96-100 25815672-7 2015 The reduction in cell death was reversed by treatment with zinc protoporphyrin, an inhibitor of HO-1, indicating that HO-1 contributed to the protective effect of EPA. Eicosapentaenoic Acid 163-166 heme oxygenase 1 Homo sapiens 118-122 25815672-8 2015 These data suggest that EPA protects against H(2)O(2)-induced oxidative stress in endothelial cells by activating Nrf2 and inducting HO-1 expression. Eicosapentaenoic Acid 24-27 heme oxygenase 1 Homo sapiens 133-137 25815672-8 2015 These data suggest that EPA protects against H(2)O(2)-induced oxidative stress in endothelial cells by activating Nrf2 and inducting HO-1 expression. Hydrogen Peroxide 45-53 heme oxygenase 1 Homo sapiens 133-137 25815690-9 2015 The results of the present study suggested that Pic may have the potential to prevent PA-induced impairment of insulin signaling and eNOS function, by inducing the expression of the anti-inflammatory and antioxidant, HO-1. 3,3',4,5'-tetrahydroxystilbene 48-51 heme oxygenase 1 Homo sapiens 217-221 25815690-9 2015 The results of the present study suggested that Pic may have the potential to prevent PA-induced impairment of insulin signaling and eNOS function, by inducing the expression of the anti-inflammatory and antioxidant, HO-1. Palmitic Acid 86-88 heme oxygenase 1 Homo sapiens 217-221 25815690-0 2015 Resveratrol analog piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 195-211 25815690-0 2015 Resveratrol analog piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. 3,3',4,5'-tetrahydroxystilbene 19-30 heme oxygenase 1 Homo sapiens 195-211 25815690-0 2015 Resveratrol analog piceatannol restores the palmitic acid-induced impairment of insulin signaling and production of endothelial nitric oxide via activation of anti-inflammatory and antioxidative heme oxygenase-1 in human endothelial cells. Palmitic Acid 44-57 heme oxygenase 1 Homo sapiens 195-211 26003724-0 2015 Ammonia-induced oxidative damage in neurons is prevented by resveratrol and lipoic acid with participation of heme oxygenase 1. Ammonia 0-7 heme oxygenase 1 Homo sapiens 110-126 26003724-0 2015 Ammonia-induced oxidative damage in neurons is prevented by resveratrol and lipoic acid with participation of heme oxygenase 1. Thioctic Acid 76-87 heme oxygenase 1 Homo sapiens 110-126 26003724-10 2015 Moreover, we showed that heme oxygenase 1 (HO1), a protein associated with protection against stress conditions, is involved in the beneficial effects of RSV and LA in cerebellar granule neurons. Resveratrol 154-157 heme oxygenase 1 Homo sapiens 25-41 26003724-10 2015 Moreover, we showed that heme oxygenase 1 (HO1), a protein associated with protection against stress conditions, is involved in the beneficial effects of RSV and LA in cerebellar granule neurons. Resveratrol 154-157 heme oxygenase 1 Homo sapiens 43-46 26132561-0 2015 Eriodictyol Protects Endothelial Cells against Oxidative Stress-Induced Cell Death through Modulating ERK/Nrf2/ARE-Dependent Heme Oxygenase-1 Expression. eriodictyol 0-11 heme oxygenase 1 Homo sapiens 125-141 26132561-5 2015 In this study, human umbilical vein endothelial cells (HUVECs) treated with eriodictyol showed the upregulation of HO-1 through extracellular-regulated kinase (ERK)/nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathways. eriodictyol 76-87 heme oxygenase 1 Homo sapiens 115-119 26132561-8 2015 These data demonstrate that eriodictyol induces ERK/Nrf2/ARE-mediated HO-1 upregulation in human endothelial cells, which is directly associated with its vascular protection against oxidative stress-related endothelial injury, and propose that targeting the upregulation of HO-1 is a promising approach for therapeutic intervention in cardiovascular disease. eriodictyol 28-39 heme oxygenase 1 Homo sapiens 70-74 26132561-8 2015 These data demonstrate that eriodictyol induces ERK/Nrf2/ARE-mediated HO-1 upregulation in human endothelial cells, which is directly associated with its vascular protection against oxidative stress-related endothelial injury, and propose that targeting the upregulation of HO-1 is a promising approach for therapeutic intervention in cardiovascular disease. eriodictyol 28-39 heme oxygenase 1 Homo sapiens 274-278 25904702-6 2015 Our results demonstrated that pretreatment of hASCs with melatonin, 100 muM for 3 h, significantly increased their proliferation and their expression of prosurvival P-Erk1/2 and P-Akt, and of antioxidative enzymes catalase and heme oxygenase (HO)-1. Melatonin 57-66 heme oxygenase 1 Homo sapiens 227-248 25978654-4 2015 In addition, NHDC induces the phase II antioxidant enzymes heme oxygenase 1 (HO-1) and NAD(P)H/quinone oxidoreductase 1 (NQO1) through the activation of the nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/antioxidant response element (ARE) signaling. neohesperidin dihydrochalcone 13-17 heme oxygenase 1 Homo sapiens 59-75 26110640-11 2015 Enhanced HO-1 expression in dh404 treated islets was confirmed by Western Blot assay. dh404 28-33 heme oxygenase 1 Homo sapiens 9-13 26045540-3 2015 HO-1 converts the pro-oxidant, proinflammatory heme molecule into metabolites with antioxidant, anti-inflammatory, and proliferative activities. Heme 47-51 heme oxygenase 1 Homo sapiens 0-4 26045540-4 2015 Previously published work has shown that KSHV-infected EC in vitro proliferate in response to free heme in a HO-1-dependent manner, thus implicating virus-enhanced HO-1 activity in KS tumorigenesis. Heme 99-103 heme oxygenase 1 Homo sapiens 109-113 26045540-4 2015 Previously published work has shown that KSHV-infected EC in vitro proliferate in response to free heme in a HO-1-dependent manner, thus implicating virus-enhanced HO-1 activity in KS tumorigenesis. Heme 99-103 heme oxygenase 1 Homo sapiens 164-168 26045540-12 2015 The detoxifying action of HO-1 is critical for the protection of cells exposed to high heme levels. Heme 87-91 heme oxygenase 1 Homo sapiens 26-30 26045540-15 2015 Our previous work showed that KSHV-infected cells proliferate in response to heme and that this occurs in a HO-1-dependent manner. Heme 77-81 heme oxygenase 1 Homo sapiens 108-112 26045540-16 2015 We therefore hypothesize that KSHV induction of HO-1 contributes to KS tumor development via heme metabolism and propose that HO-1 be evaluated as a therapeutic target for KS. Heme 93-97 heme oxygenase 1 Homo sapiens 48-52 25932745-8 2015 Conversely, lycopene treatment enhanced heme oxygenase-1 (HO-1) gene expression through the activation of phosphoinositide 3-kinase (PI3K)/Akt pathway, followed by induction of the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation; in addition, HO-1 silencing partially inhibited the repressive effects of lycopene on strain-induced ET-1 expression. Lycopene 12-20 heme oxygenase 1 Homo sapiens 268-272 25615876-0 2015 Pretreatment with pPolyHb attenuates H2O2-induced endothelial cell injury through inhibition of JNK/p38 MAPK pathway by upregulation of heme oxygenase-1. Hydrogen Peroxide 37-41 heme oxygenase 1 Homo sapiens 136-152 25615876-3 2015 The results showed that the pretreatment augmented heme oxygenase-1 (HO-1) expression, and at the same time, decreased the phosphorylation of JNK/p38 mitogen-activated protein kinase (MAPK) and intracellular ROS generation in H2O2-treated HUVECs. ros 208-211 heme oxygenase 1 Homo sapiens 51-67 25615876-3 2015 The results showed that the pretreatment augmented heme oxygenase-1 (HO-1) expression, and at the same time, decreased the phosphorylation of JNK/p38 mitogen-activated protein kinase (MAPK) and intracellular ROS generation in H2O2-treated HUVECs. ros 208-211 heme oxygenase 1 Homo sapiens 69-73 25615876-3 2015 The results showed that the pretreatment augmented heme oxygenase-1 (HO-1) expression, and at the same time, decreased the phosphorylation of JNK/p38 mitogen-activated protein kinase (MAPK) and intracellular ROS generation in H2O2-treated HUVECs. Hydrogen Peroxide 226-230 heme oxygenase 1 Homo sapiens 51-67 25615876-3 2015 The results showed that the pretreatment augmented heme oxygenase-1 (HO-1) expression, and at the same time, decreased the phosphorylation of JNK/p38 mitogen-activated protein kinase (MAPK) and intracellular ROS generation in H2O2-treated HUVECs. Hydrogen Peroxide 226-230 heme oxygenase 1 Homo sapiens 69-73 25615876-4 2015 Moreover, the inhibition of HO-1 expression by tin porphyrin (SnPP) abolished the protective effects of pPolyHb, which suggested that the cytoprotective effect of pPolyHb involves upregulating HO-1 and subsequently decreasing the phosphorylation of the JNK and p38 MAPK and ROS generation. tin porphyrin 47-60 heme oxygenase 1 Homo sapiens 28-32 25615876-4 2015 Moreover, the inhibition of HO-1 expression by tin porphyrin (SnPP) abolished the protective effects of pPolyHb, which suggested that the cytoprotective effect of pPolyHb involves upregulating HO-1 and subsequently decreasing the phosphorylation of the JNK and p38 MAPK and ROS generation. tin porphyrin 47-60 heme oxygenase 1 Homo sapiens 193-197 25615876-4 2015 Moreover, the inhibition of HO-1 expression by tin porphyrin (SnPP) abolished the protective effects of pPolyHb, which suggested that the cytoprotective effect of pPolyHb involves upregulating HO-1 and subsequently decreasing the phosphorylation of the JNK and p38 MAPK and ROS generation. S-Nitroso-N-propionyl-D,L-penicillamine 62-66 heme oxygenase 1 Homo sapiens 28-32 25615876-4 2015 Moreover, the inhibition of HO-1 expression by tin porphyrin (SnPP) abolished the protective effects of pPolyHb, which suggested that the cytoprotective effect of pPolyHb involves upregulating HO-1 and subsequently decreasing the phosphorylation of the JNK and p38 MAPK and ROS generation. S-Nitroso-N-propionyl-D,L-penicillamine 62-66 heme oxygenase 1 Homo sapiens 193-197 25615876-4 2015 Moreover, the inhibition of HO-1 expression by tin porphyrin (SnPP) abolished the protective effects of pPolyHb, which suggested that the cytoprotective effect of pPolyHb involves upregulating HO-1 and subsequently decreasing the phosphorylation of the JNK and p38 MAPK and ROS generation. ros 274-277 heme oxygenase 1 Homo sapiens 28-32 25932745-0 2015 Lycopene inhibits cyclic strain-induced endothelin-1 expression through the suppression of reactive oxygen species generation and induction of heme oxygenase-1 in human umbilical vein endothelial cells. Lycopene 0-8 heme oxygenase 1 Homo sapiens 143-159 25932745-8 2015 Conversely, lycopene treatment enhanced heme oxygenase-1 (HO-1) gene expression through the activation of phosphoinositide 3-kinase (PI3K)/Akt pathway, followed by induction of the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation; in addition, HO-1 silencing partially inhibited the repressive effects of lycopene on strain-induced ET-1 expression. Lycopene 12-20 heme oxygenase 1 Homo sapiens 40-56 25932745-8 2015 Conversely, lycopene treatment enhanced heme oxygenase-1 (HO-1) gene expression through the activation of phosphoinositide 3-kinase (PI3K)/Akt pathway, followed by induction of the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation; in addition, HO-1 silencing partially inhibited the repressive effects of lycopene on strain-induced ET-1 expression. Lycopene 12-20 heme oxygenase 1 Homo sapiens 58-62 25932745-8 2015 Conversely, lycopene treatment enhanced heme oxygenase-1 (HO-1) gene expression through the activation of phosphoinositide 3-kinase (PI3K)/Akt pathway, followed by induction of the nuclear factor erythroid 2-related factor 2 (Nrf2) nuclear translocation; in addition, HO-1 silencing partially inhibited the repressive effects of lycopene on strain-induced ET-1 expression. Lycopene 329-337 heme oxygenase 1 Homo sapiens 58-62 25932745-9 2015 In summary, our study showed, for the first time, that lycopene inhibits cyclic strain-induced ET-1 gene expression through the suppression of ROS generation and induction of HO-1 in HUVECs. Lycopene 55-63 heme oxygenase 1 Homo sapiens 175-179 26125799-1 2015 We evaluated the effects of down-regulated heme oxygenase (HO)-1 expression on the proliferation of the acute myelocytic leukemia Kasumi-1 cell line by using the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) in combination with daunorubicin (DNR), and evaluated the mechanism. zinc protoporphyrin 177-199 heme oxygenase 1 Homo sapiens 43-64 26125799-1 2015 We evaluated the effects of down-regulated heme oxygenase (HO)-1 expression on the proliferation of the acute myelocytic leukemia Kasumi-1 cell line by using the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX) in combination with daunorubicin (DNR), and evaluated the mechanism. Daunorubicin 229-241 heme oxygenase 1 Homo sapiens 43-64 25714596-0 2015 Impact of Enhanced Production of Endogenous Heme Oxygenase-1 by Pitavastatin on Survival and Functional Activities of Bone Marrow-derived Mesenchymal Stem Cells. pitavastatin 64-76 heme oxygenase 1 Homo sapiens 44-60 25714596-3 2015 We investigated the protective effect of statin-induced overexpression of HO-1 by examining changes in gene expression and function in MSCs after pitavastatin treatment. pitavastatin 146-158 heme oxygenase 1 Homo sapiens 74-78 25714596-4 2015 The relative expression of the HO-1 and endothelial nitric oxide synthase genes in MSCs was significantly increased after treatment with pitavastatin (MSCs). pitavastatin 137-149 heme oxygenase 1 Homo sapiens 31-35 25714596-9 2015 These results demonstrate that pitavastatin can enhance endogenous HO-1 expression in MSCs, which may protect the cells into the environment of oxidative stress with partial activation of endothelial nitric oxide synthase and Akt phosphorylation. pitavastatin 31-43 heme oxygenase 1 Homo sapiens 67-71 25956277-1 2015 HO-1 gene encodes heme oxygenase-1 enzyme that catalyzes the oxidation of heme to carbon monoxide (CO). Heme 18-22 heme oxygenase 1 Homo sapiens 0-4 26072076-0 2015 Protective effect of HO-1 transfection against ethanol-induced osteoblast damage. Ethanol 47-54 heme oxygenase 1 Homo sapiens 21-25 26072076-2 2015 The aim of this study was to observe if HO-1 transfection could inhibit the damage of osteoblasts induced by ethanol. Ethanol 109-116 heme oxygenase 1 Homo sapiens 40-44 26072076-8 2015 These results suggest that HO-1 plays a protective role in osteoblasts, and HO-1 transfection can effectively inhibit bone damage induced by ethanol. Ethanol 141-148 heme oxygenase 1 Homo sapiens 76-80 25765514-8 2015 Among them, Hmox1, Bdkrb2, Adra2b, Ptgs2, Col1a2 and Tbxa2r are associated with endothelium-derived hyperpolarizing factor (EDHF). endothelium-derived hyperpolarizing factor 80-122 heme oxygenase 1 Homo sapiens 12-17 25765514-8 2015 Among them, Hmox1, Bdkrb2, Adra2b, Ptgs2, Col1a2 and Tbxa2r are associated with endothelium-derived hyperpolarizing factor (EDHF). edhf 124-128 heme oxygenase 1 Homo sapiens 12-17 25776458-0 2015 Myrosinase-treated glucoerucin is a potent inducer of the Nrf2 target gene heme oxygenase 1--studies in cultured HT-29 cells and mice. glucoerucin 19-30 heme oxygenase 1 Homo sapiens 75-91 25956277-1 2015 HO-1 gene encodes heme oxygenase-1 enzyme that catalyzes the oxidation of heme to carbon monoxide (CO). Carbon Monoxide 82-97 heme oxygenase 1 Homo sapiens 0-4 25633656-6 2015 The results of the present study demonstrated that the mRNA expression levels of HO-1 increased in a dose-dependent manner in the HepG2.2.15 cells, following exposure to 5-50 microM hemin. Hemin 182-187 heme oxygenase 1 Homo sapiens 81-85 25751573-1 2015 BACKGROUND: Heme oxygenase 1 (HO1) catalyzes heme degradation, and offers protection for several organs, including the kidney. Heme 45-49 heme oxygenase 1 Homo sapiens 12-28 25751573-1 2015 BACKGROUND: Heme oxygenase 1 (HO1) catalyzes heme degradation, and offers protection for several organs, including the kidney. Heme 45-49 heme oxygenase 1 Homo sapiens 30-33 25956277-1 2015 HO-1 gene encodes heme oxygenase-1 enzyme that catalyzes the oxidation of heme to carbon monoxide (CO). Carbon Monoxide 82-97 heme oxygenase 1 Homo sapiens 18-34 25956277-1 2015 HO-1 gene encodes heme oxygenase-1 enzyme that catalyzes the oxidation of heme to carbon monoxide (CO). Carbon Monoxide 99-101 heme oxygenase 1 Homo sapiens 0-4 25956277-1 2015 HO-1 gene encodes heme oxygenase-1 enzyme that catalyzes the oxidation of heme to carbon monoxide (CO). Carbon Monoxide 99-101 heme oxygenase 1 Homo sapiens 18-34 25956277-3 2015 A (GT) n dinucleotide repeat at HO-1 promoter is a polymorphic region and modulates gene transcription and associated with some of diseases. Dinucleoside Phosphates 9-21 heme oxygenase 1 Homo sapiens 32-36 25849895-2 2015 Mammals contain two isoforms of this enzyme, HO2 and HO1, which share the same alpha-helical fold forming the catalytic core and heme binding site, as well as a membrane spanning helix at their C-termini. Heme 129-133 heme oxygenase 1 Homo sapiens 53-56 25802333-0 2015 Induction of heme oxygenase-1 by Na+-H+ exchanger 1 protein plays a crucial role in imatinib-resistant chronic myeloid leukemia cells. Imatinib Mesylate 84-92 heme oxygenase 1 Homo sapiens 13-29 25802333-3 2015 After treatment with the specific NHE1 inhibitor cariporide to decrease intracellular pH (pHi), the heme oxygenase-1 (HO-1) levels of the K562R cell line and cells from IM-insensitive CML patients decreased. cariporide 49-59 heme oxygenase 1 Homo sapiens 100-116 26078725-10 2015 ROS scavenger N-acetyl-L-cysteine (NAC) blocked the autophagy process induced by 5cGy alpha particle, the upregulation of Nrf2 and HO-1, as well as the induced radio-resistance. ros 0-3 heme oxygenase 1 Homo sapiens 131-135 26078725-11 2015 In conclusion, ROS elevation caused by LDIR promoted Autophagy/Nrf2-HO-1 and conferred radio-resistance in A549. ros 15-18 heme oxygenase 1 Homo sapiens 68-72 25891083-13 2015 CONCLUSION: Curcumin inhibits appoptosin-induced apoptosis in SH-SY5Y cells by upregulating the expression of HO-1, reducing the production of intracellular heme and ROS, and preventing the DeltaPsim loss. Curcumin 12-20 heme oxygenase 1 Homo sapiens 110-114 25941985-0 2015 Camptothecin suppresses expression of matrix metalloproteinase-9 and vascular endothelial growth factor in DU145 cells through PI3K/Akt-mediated inhibition of NF-kappaB activity and Nrf2-dependent induction of HO-1 expression. Camptothecin 0-12 heme oxygenase 1 Homo sapiens 210-214 25795137-7 2015 Excess intracellular ROS induced by Dex significantly suppressed the expression levels of Nrf2 and the downstream effectors, HO1 and NQO1, but these changes could be reversed by I3C. ros 21-24 heme oxygenase 1 Homo sapiens 125-128 25795137-7 2015 Excess intracellular ROS induced by Dex significantly suppressed the expression levels of Nrf2 and the downstream effectors, HO1 and NQO1, but these changes could be reversed by I3C. Dexamethasone 36-39 heme oxygenase 1 Homo sapiens 125-128 25891083-0 2015 Curcumin inhibits appoptosin-induced apoptosis via upregulating heme oxygenase-1 expression in SH-SY5Y cells. Curcumin 0-8 heme oxygenase 1 Homo sapiens 64-80 25631293-6 2015 Moreover, DHEA stimulated the activation of Nrf2, which led to the expression of an antioxidant response gene, HO-1. Dehydroepiandrosterone 10-14 heme oxygenase 1 Homo sapiens 111-115 25941985-6 2015 We further confirmed that CPT inhibits PMA-induced MMP-9 and VEGF expression by upregulating nuclear factor-erythroid related factor-2 (Nrf2)-mediated heme oxygenase-1 (HO-1) induction. Tetradecanoylphorbol Acetate 39-42 heme oxygenase 1 Homo sapiens 151-167 25941985-6 2015 We further confirmed that CPT inhibits PMA-induced MMP-9 and VEGF expression by upregulating nuclear factor-erythroid related factor-2 (Nrf2)-mediated heme oxygenase-1 (HO-1) induction. Tetradecanoylphorbol Acetate 39-42 heme oxygenase 1 Homo sapiens 169-173 25692285-0 2015 Anti-Inflammatory Activity of Butein and Luteolin Through Suppression of NFkappaB Activation and Induction of Heme Oxygenase-1. butein 30-36 heme oxygenase 1 Homo sapiens 110-126 25744070-11 2015 Inhibition of HO-1 by tin protoporphyrin IX exacerbated autophagy and mitochondrial activity as well as cell viability in young cells. tin protoporphyrin IX 22-43 heme oxygenase 1 Homo sapiens 14-18 25303683-4 2015 At 20% O2, DMF induced a stronger antioxidant response compared to 5% O2 as evidenced by a higher expression of heme oxygenase-1, NAD(P)H:quinone oxydoreductase-1 and superoxide dismutase-2. Dimethyl Fumarate 11-14 heme oxygenase 1 Homo sapiens 112-189 25303683-4 2015 At 20% O2, DMF induced a stronger antioxidant response compared to 5% O2 as evidenced by a higher expression of heme oxygenase-1, NAD(P)H:quinone oxydoreductase-1 and superoxide dismutase-2. Oxygen 70-72 heme oxygenase 1 Homo sapiens 112-189 25692285-7 2015 Additionally, the anti-inflammatory activities of butein and luteolin involved the induction of HO-1 expression, as confirmed by the zinc protoporphyrin (ZnPP) treatment (HO-1 selective inhibitor) and HO-1 small interfering (si)RNA system. zinc protoporphyrin 133-152 heme oxygenase 1 Homo sapiens 96-100 25692285-7 2015 Additionally, the anti-inflammatory activities of butein and luteolin involved the induction of HO-1 expression, as confirmed by the zinc protoporphyrin (ZnPP) treatment (HO-1 selective inhibitor) and HO-1 small interfering (si)RNA system. zinc protoporphyrin 154-158 heme oxygenase 1 Homo sapiens 96-100 25771143-0 2015 Rosuvastatin suppresses atrial tachycardia-induced cellular remodeling via Akt/Nrf2/heme oxygenase-1 pathway. Rosuvastatin Calcium 0-12 heme oxygenase 1 Homo sapiens 84-100 25692285-8 2015 ZnPP-mediated downregulation and siRNA-mediated knockdown of HO-1 significantly abolished the inhibitory effects of butein and luteolin on the production of NO in LPS-induced macrophages. butein 116-122 heme oxygenase 1 Homo sapiens 61-65 25771143-4 2015 Treatment of cultured atrium-derived myocytes (HL-1 cell line) with rosuvastatin enhanced HO-1 expression/activity and attenuated tachypacing-induced oxidative stress and myofibril degradation. Rosuvastatin Calcium 68-80 heme oxygenase 1 Homo sapiens 90-94 25620054-5 2015 HO-1 expression and/or activity were inhibited by siRNA or tin protoporphyrin (Sn PPIX) and enhanced by an expression plasmid or cobalt protoporphyrin (CoPPIX). tin protoporphyrin IX 59-77 heme oxygenase 1 Homo sapiens 0-4 25771143-5 2015 Heme oxygenase-1 inhibitors and small-interfering RNA for HO-1 blocked the inhibitory effect of rosuvastatin on tachypacing-stimulated changes, suggesting the crucial role of HO-1 in mediating the effect of rosuvastatin. Rosuvastatin Calcium 96-108 heme oxygenase 1 Homo sapiens 58-62 25771143-5 2015 Heme oxygenase-1 inhibitors and small-interfering RNA for HO-1 blocked the inhibitory effect of rosuvastatin on tachypacing-stimulated changes, suggesting the crucial role of HO-1 in mediating the effect of rosuvastatin. Rosuvastatin Calcium 96-108 heme oxygenase 1 Homo sapiens 175-179 25771143-5 2015 Heme oxygenase-1 inhibitors and small-interfering RNA for HO-1 blocked the inhibitory effect of rosuvastatin on tachypacing-stimulated changes, suggesting the crucial role of HO-1 in mediating the effect of rosuvastatin. Rosuvastatin Calcium 207-219 heme oxygenase 1 Homo sapiens 58-62 25771143-5 2015 Heme oxygenase-1 inhibitors and small-interfering RNA for HO-1 blocked the inhibitory effect of rosuvastatin on tachypacing-stimulated changes, suggesting the crucial role of HO-1 in mediating the effect of rosuvastatin. Rosuvastatin Calcium 207-219 heme oxygenase 1 Homo sapiens 175-179 25771143-7 2015 Furthermore, the involvement of Akt/Nrf2/HO-1 pathway in the antioxidant effect of rosuvastatin was documented in an ex vivo tachypacing model. Rosuvastatin Calcium 83-95 heme oxygenase 1 Homo sapiens 41-45 25843086-3 2015 In this study, we found that the expression levels of Nrf2 and its target genes GCLC, HO-1, NQO1 were significantly higher in cisplatin-resistant A549 (A549/CDDP) cells than those in A549 cells, and this resistance was partially reversed by Nrf2 siRNA. Cisplatin 126-135 heme oxygenase 1 Homo sapiens 86-90 25896339-4 2015 Following treatment with quercetin, analyses of the nuclear level of Nrf2, Nrf2 antioxidant response element-binding assay, Nrf2 promoter-luc assay, and RT-PCR toward the Nrf2-regulated gene, heme oxygenase-1, demonstrated that the induced Nrf2 is transcriptionally active. Quercetin 25-34 heme oxygenase 1 Homo sapiens 192-208 25620054-5 2015 HO-1 expression and/or activity were inhibited by siRNA or tin protoporphyrin (Sn PPIX) and enhanced by an expression plasmid or cobalt protoporphyrin (CoPPIX). tin protoporphyrin IX 79-86 heme oxygenase 1 Homo sapiens 0-4 25620054-5 2015 HO-1 expression and/or activity were inhibited by siRNA or tin protoporphyrin (Sn PPIX) and enhanced by an expression plasmid or cobalt protoporphyrin (CoPPIX). cobaltiprotoporphyrin 129-150 heme oxygenase 1 Homo sapiens 0-4 25620054-5 2015 HO-1 expression and/or activity were inhibited by siRNA or tin protoporphyrin (Sn PPIX) and enhanced by an expression plasmid or cobalt protoporphyrin (CoPPIX). coppix 152-158 heme oxygenase 1 Homo sapiens 0-4 25620054-7 2015 Inhibition of HO-1 enzymatic activity with SnPPIX and silencing of the HO-1 gene by siRNA enhanced DEP-induced ROS production, further decreased cell viability and increased expression of inflammatory and cell adhesion molecules. tin protoporphyrin IX 43-49 heme oxygenase 1 Homo sapiens 14-18 25620054-7 2015 Inhibition of HO-1 enzymatic activity with SnPPIX and silencing of the HO-1 gene by siRNA enhanced DEP-induced ROS production, further decreased cell viability and increased expression of inflammatory and cell adhesion molecules. ros 111-114 heme oxygenase 1 Homo sapiens 14-18 25620054-7 2015 Inhibition of HO-1 enzymatic activity with SnPPIX and silencing of the HO-1 gene by siRNA enhanced DEP-induced ROS production, further decreased cell viability and increased expression of inflammatory and cell adhesion molecules. ros 111-114 heme oxygenase 1 Homo sapiens 71-75 25620054-8 2015 On the other hand, overexpression of the HO-1 gene by a pcDNA 3.1D/V5-HO-1 plasmid significantly mitigated ROS production, increased cell survival and decreased the expression of inflammatory genes. ros 107-110 heme oxygenase 1 Homo sapiens 41-45 25620054-8 2015 On the other hand, overexpression of the HO-1 gene by a pcDNA 3.1D/V5-HO-1 plasmid significantly mitigated ROS production, increased cell survival and decreased the expression of inflammatory genes. ros 107-110 heme oxygenase 1 Homo sapiens 70-74 25714994-5 2015 Ziram increased intracellular levels of zinc and copper, DMDC increased intracellular levels of only copper, and both dithiocarbamates enhanced oxidative injury evidenced by elevated levels of protein carbonyls and expression of heme oxygenase-1. dithiocarbamates 118-134 heme oxygenase 1 Homo sapiens 229-245 24931165-1 2015 Heme oxygenase-1 (HO-1) is a heme-degrading enzyme anchored in the endoplasmic reticulum by a carboxyl-terminal transmembrane segment (TMS). Heme 29-33 heme oxygenase 1 Homo sapiens 0-16 24931165-1 2015 Heme oxygenase-1 (HO-1) is a heme-degrading enzyme anchored in the endoplasmic reticulum by a carboxyl-terminal transmembrane segment (TMS). Heme 29-33 heme oxygenase 1 Homo sapiens 18-22 25742418-5 2015 After translocation, Nrf2 subsequently binds to the antioxidant response element (ARE), up-regulated heme oxygenase-1 (HO-1), and NADH quinone oxidoreductase subunit 1 (NQO1), which may be considered as an antioxidative response to TCBQ-intoxication. tetrachlorobenzoquinone 232-236 heme oxygenase 1 Homo sapiens 101-117 25875041-4 2015 The aim of the present study was to investigate whether EGCG could also inhibit PM2.5-induced oxidative stress by activating the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in human umbilical vein endothelial cells (HUVECs). epigallocatechin gallate 56-60 heme oxygenase 1 Homo sapiens 171-187 25652453-0 2015 5-aminolevulinic acid combined with sodium ferrous citrate ameliorates H2O2-induced cardiomyocyte hypertrophy via activation of the MAPK/Nrf2/HO-1 pathway. 5-amino levulinic acid 0-21 heme oxygenase 1 Homo sapiens 142-146 25652453-0 2015 5-aminolevulinic acid combined with sodium ferrous citrate ameliorates H2O2-induced cardiomyocyte hypertrophy via activation of the MAPK/Nrf2/HO-1 pathway. Ferromia 36-58 heme oxygenase 1 Homo sapiens 142-146 25652453-0 2015 5-aminolevulinic acid combined with sodium ferrous citrate ameliorates H2O2-induced cardiomyocyte hypertrophy via activation of the MAPK/Nrf2/HO-1 pathway. Hydrogen Peroxide 71-75 heme oxygenase 1 Homo sapiens 142-146 25652453-3 2015 In this study, we investigated whether the heme precursor 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC) could protect cardiomyocytes from H2O2-induced hypertrophy via modulation of HO-1 expression. Heme 43-47 heme oxygenase 1 Homo sapiens 199-203 25652453-3 2015 In this study, we investigated whether the heme precursor 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC) could protect cardiomyocytes from H2O2-induced hypertrophy via modulation of HO-1 expression. 5-amino levulinic acid 58-79 heme oxygenase 1 Homo sapiens 199-203 25652453-3 2015 In this study, we investigated whether the heme precursor 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC) could protect cardiomyocytes from H2O2-induced hypertrophy via modulation of HO-1 expression. 5-amino levulinic acid 81-86 heme oxygenase 1 Homo sapiens 199-203 25652453-3 2015 In this study, we investigated whether the heme precursor 5-aminolevulinic acid (5-ALA) with sodium ferrous citrate (SFC) could protect cardiomyocytes from H2O2-induced hypertrophy via modulation of HO-1 expression. SFC 117-120 heme oxygenase 1 Homo sapiens 199-203 25652453-13 2015 3) ERK1/2, p38, and SAPK/JNK signaling pathways were activated and modulate 5-ALA- and SFC-enhanced HO-1 expression. 5-amino levulinic acid 76-81 heme oxygenase 1 Homo sapiens 100-104 25652453-15 2015 In conclusion, our data suggest that 5-ALA and SFC protect HL-1 cells from H2O2-induced cardiac hypertrophy via activation of the MAPK/Nrf2/HO-1 signaling pathway. 5-amino levulinic acid 37-42 heme oxygenase 1 Homo sapiens 140-144 25652453-15 2015 In conclusion, our data suggest that 5-ALA and SFC protect HL-1 cells from H2O2-induced cardiac hypertrophy via activation of the MAPK/Nrf2/HO-1 signaling pathway. Hydrogen Peroxide 75-79 heme oxygenase 1 Homo sapiens 140-144 25875041-8 2015 In addition, silencing Nrf2 abolished EGCG-induced Nrf2 and HO-1 upregulation and enhancement of cell viability. epigallocatechin gallate 38-42 heme oxygenase 1 Homo sapiens 60-64 25875041-9 2015 The present study suggests that EGCG protects HUVECs from PM2.5-induced oxidative stress injury by upregulating Nrf2/HO-1 via activation of the p38 MAPK and the ERK1/2 signaling pathways. epigallocatechin gallate 32-36 heme oxygenase 1 Homo sapiens 117-121 25875041-4 2015 The aim of the present study was to investigate whether EGCG could also inhibit PM2.5-induced oxidative stress by activating the nuclear factor E2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway in human umbilical vein endothelial cells (HUVECs). epigallocatechin gallate 56-60 heme oxygenase 1 Homo sapiens 189-193 25875041-6 2015 Western blotting and PCR demonstrated that EGCG increased Nrf2 and HO-1 expression in HUVECs that had been exposed to PM2.5. epigallocatechin gallate 43-47 heme oxygenase 1 Homo sapiens 67-71 28962395-8 2015 Our present experimental data support the notion that Cr(VI) caused mitochondrial damage, apoptosis, oxidative stress, and subsequently lead to a strong induction of HO1, GCLC and SOD2 via the Nrf-2 signaling pathway in hepatocytes. Chromium 54-56 heme oxygenase 1 Homo sapiens 166-169 25807407-7 2015 RESULTS: Ketamine treatment dose-dependently attenuated the increased levels of proinflammatory mediators (HMGB1, nitric oxide, tumor necrosis factor alpha, and interleukin 1beta) and increased the HO-1 protein expression in LPS-activated macrophages. Ketamine 9-17 heme oxygenase 1 Homo sapiens 198-202 25835394-0 2015 Heme oxygenase-1 protects corexit 9500A-induced respiratory epithelial injury across species. corexit 26-33 heme oxygenase 1 Homo sapiens 0-16 25835394-5 2015 CE induced the expression of HO-1 as well as C-reactive protein (CRP) and NADPH oxidase 4 (NOX4), which are associated with ROS production. ros 124-127 heme oxygenase 1 Homo sapiens 29-33 25835394-9 2015 Treatment with carbon monoxide releasing molecule-2 (CORM-2) significantly inhibited CE-induced ROS production, while the addition of HO-1 inhibitor, significantly increased CE-induced ROS production and apoptosis, suggesting a protective role of HO-1 or its reaction product, CO, in CE-induced apoptosis. Carbon Monoxide 15-30 heme oxygenase 1 Homo sapiens 247-251 25835394-9 2015 Treatment with carbon monoxide releasing molecule-2 (CORM-2) significantly inhibited CE-induced ROS production, while the addition of HO-1 inhibitor, significantly increased CE-induced ROS production and apoptosis, suggesting a protective role of HO-1 or its reaction product, CO, in CE-induced apoptosis. ros 185-188 heme oxygenase 1 Homo sapiens 134-138 25377066-6 2015 Moreover, we have revealed the PB treatment resulted in an increase in nuclear factor E2-related factor 2 (Nrf2) protein levels and subsequent activation of antioxidant response element (ARE) pathway genes of heme oxygenase-1 (HO-1) and gamma-glutamylcysteine synthetase (gamma-GCS) in SH-SY5Y cells. pinocembrin 31-33 heme oxygenase 1 Homo sapiens 209-225 25595329-3 2015 It has been recently suggested that the heme-catabolizing enzyme heme oxygenase-1 is an essential component of the mesenchymal stromal cell-driven immune suppressive response. Heme 40-44 heme oxygenase 1 Homo sapiens 65-81 25595329-4 2015 Because mesenchymal stromal cells upregulate indoleamine 2,3-dioxygenase expression on interferon-gamma priming and indoleamine 2,3-dioxygenase requires heme as a cofactor for optimal catabolic function, we investigated the potential antagonism of heme oxygenase-1 activity on indoleamine 2, 3-dioxygenase and the impact on mesenchymal stromal cell immune plasticity. Heme 153-157 heme oxygenase 1 Homo sapiens 248-264 25667213-0 2015 Sofalcone upregulates the nuclear factor (erythroid-derived 2)-like 2/heme oxygenase-1 pathway, reduces soluble fms-like tyrosine kinase-1, and quenches endothelial dysfunction: potential therapeutic for preeclampsia. sofalcone 0-9 heme oxygenase 1 Homo sapiens 70-86 25667213-5 2015 Sofalcone, a gastric antiulcer agent in clinical use, is known to induce HO-1 in gastric epithelium. sofalcone 0-9 heme oxygenase 1 Homo sapiens 73-77 25667213-6 2015 We aimed to investigate whether sofalcone induces HO-1 and reduces sFlt-1 release from primary human placental and endothelial cells and blocks endothelial dysfunction in vitro. sofalcone 32-41 heme oxygenase 1 Homo sapiens 50-54 25667213-9 2015 Sofalcone potently increased HO-1 mRNA and protein in both primary trophoblasts and human umbilical vein endothelial cells. sofalcone 0-9 heme oxygenase 1 Homo sapiens 29-33 25667213-13 2015 These results indicate that in primary human tissues, sofalcone can potently activate antioxidant nuclear factor (erythroid-derived 2)-like 2/HO-1 pathway, decrease sFlt-1 production, and ameliorate endothelial dysfunction. sofalcone 54-63 heme oxygenase 1 Homo sapiens 142-146 25653454-9 2015 The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. Protoporphyrin IX cobalt chloride 44-77 heme oxygenase 1 Homo sapiens 26-30 25653454-9 2015 The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. Protoporphyrin IX cobalt chloride 44-77 heme oxygenase 1 Homo sapiens 108-112 25653454-9 2015 The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. COPP protocol 79-83 heme oxygenase 1 Homo sapiens 26-30 25653454-9 2015 The suppressive effect of HO-1 induction by protoporphyrin IX cobalt chloride (CoPP; a classical inducer of HO-1 expression) on PRRSV replication in MARC-145 cells and primary porcine alveolar macrophages could also be reversed by overexpression of miR-24-3p. COPP protocol 79-83 heme oxygenase 1 Homo sapiens 108-112 25652099-0 2015 Downregulation of HO-1 promoted apoptosis induced by decitabine via increasing p15INK4B promoter demethylation in myelodysplastic syndrome. Decitabine 53-63 heme oxygenase 1 Homo sapiens 18-22 25652099-2 2015 In this study, heme oxygenase-1 (HO-1) was overexpressed in MDS cell line SKM-1, which was closely related to resistance to decitabine-induced apoptosis. Decitabine 124-134 heme oxygenase 1 Homo sapiens 15-31 25652099-2 2015 In this study, heme oxygenase-1 (HO-1) was overexpressed in MDS cell line SKM-1, which was closely related to resistance to decitabine-induced apoptosis. Decitabine 124-134 heme oxygenase 1 Homo sapiens 33-37 25652099-3 2015 We aimed to further investigate the role of HO-1 in apoptosis induced by low-dose decitabine in SKM-1 cells. Decitabine 82-92 heme oxygenase 1 Homo sapiens 44-48 25652099-5 2015 In contrast, downregulation of HO-1 enhanced decitabine-induced apoptosis but reduced accumulation of the S phase in cell cycle. Decitabine 45-55 heme oxygenase 1 Homo sapiens 31-35 25652099-12 2015 In conclusion, overexpression of HO-1 indicated resistance to demethylation of p15(INK4B) induced by decitabine. Decitabine 101-111 heme oxygenase 1 Homo sapiens 33-37 25807407-0 2015 Ketamine reduces LPS-induced HMGB1 via activation of the Nrf2/HO-1 pathway and NF-kappaB suppression. Ketamine 0-8 heme oxygenase 1 Homo sapiens 62-66 25807407-9 2015 In addition, the present study also demonstrated that ketamine induced HO-1 expression through the nuclear translocation of nuclear factor E2-related factor 2 in macrophages. Ketamine 54-62 heme oxygenase 1 Homo sapiens 71-75 25807407-11 2015 CONCLUSION: Ketamine inhibits the release of HMGB1 in LPS-stimulated macrophages, and this effect is at least partly mediated by the activation of the Nrf2/HO-1 pathway and NF-kappaB suppression. Ketamine 12-20 heme oxygenase 1 Homo sapiens 156-160 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Heme 95-99 heme oxygenase 1 Homo sapiens 0-16 25813625-9 2015 Taken together, these findings suggest that direct interaction of piperlongumine with Keap1 leads to the upregulation of Nrf2-mediated HO-1 expression, and HO-1 determines the differential response of breast normal cells and cancer cells to piperlongumine. piperlonguminine 66-80 heme oxygenase 1 Homo sapiens 135-139 25813625-9 2015 Taken together, these findings suggest that direct interaction of piperlongumine with Keap1 leads to the upregulation of Nrf2-mediated HO-1 expression, and HO-1 determines the differential response of breast normal cells and cancer cells to piperlongumine. piperlonguminine 241-255 heme oxygenase 1 Homo sapiens 156-160 25891728-6 2015 Immunohistochemistry and Western blot analysis data revealed that the FK506 treatment increased expression of heme oxygenase-1 (HO-1) in Jurkat cells in a dose-dependent manner. Tacrolimus 70-75 heme oxygenase 1 Homo sapiens 110-126 25891728-6 2015 Immunohistochemistry and Western blot analysis data revealed that the FK506 treatment increased expression of heme oxygenase-1 (HO-1) in Jurkat cells in a dose-dependent manner. Tacrolimus 70-75 heme oxygenase 1 Homo sapiens 128-132 25891728-7 2015 HO-1 expression was induced after 6 hours of treatment of FK506 to Jurkat cells, peaked at 24 hours, and then decreased after 48 hours. Tacrolimus 58-63 heme oxygenase 1 Homo sapiens 0-4 25891728-8 2015 CONCLUSIONS: These results suggest that FK506 induces Nrf 2-driven transcriptional activation of the antioxidant response element by activating HO-1 and free radicals such as reactive oxygen species and nitric oxide. Tacrolimus 40-45 heme oxygenase 1 Homo sapiens 144-148 25891728-8 2015 CONCLUSIONS: These results suggest that FK506 induces Nrf 2-driven transcriptional activation of the antioxidant response element by activating HO-1 and free radicals such as reactive oxygen species and nitric oxide. Reactive Oxygen Species 175-198 heme oxygenase 1 Homo sapiens 144-148 25891728-8 2015 CONCLUSIONS: These results suggest that FK506 induces Nrf 2-driven transcriptional activation of the antioxidant response element by activating HO-1 and free radicals such as reactive oxygen species and nitric oxide. Nitric Oxide 203-215 heme oxygenase 1 Homo sapiens 144-148 25813625-0 2015 Heme oxygenase-1 determines the differential response of breast cancer and normal cells to piperlongumine. piperlonguminine 91-105 heme oxygenase 1 Homo sapiens 0-16 25813625-4 2015 Interestingly, this opposing effect of piperlongumine appears to be mediated by heme oxygenase-1 (HO-1). piperlonguminine 39-53 heme oxygenase 1 Homo sapiens 80-96 25813625-4 2015 Interestingly, this opposing effect of piperlongumine appears to be mediated by heme oxygenase-1 (HO-1). piperlonguminine 39-53 heme oxygenase 1 Homo sapiens 98-102 25813625-5 2015 Piperlongumine upregulated HO-1 expression through the activation of nuclear factor-erythroid-2-related factor-2 (Nrf2) signaling in both MCF-7 and MCF-10A cells. piperlonguminine 0-14 heme oxygenase 1 Homo sapiens 27-31 25813625-6 2015 However, knockdown of HO-1 expression and pharmacological inhibition of its activity abolished the ability of piperlongumine to induce apoptosis in MCF-7 cells, whereas those promoted apoptosis in MCF-10A cells, indicating that HO-1 has anti-tumor functions in cancer cells but cytoprotective functions in normal cells. piperlonguminine 110-124 heme oxygenase 1 Homo sapiens 22-26 25813625-6 2015 However, knockdown of HO-1 expression and pharmacological inhibition of its activity abolished the ability of piperlongumine to induce apoptosis in MCF-7 cells, whereas those promoted apoptosis in MCF-10A cells, indicating that HO-1 has anti-tumor functions in cancer cells but cytoprotective functions in normal cells. piperlonguminine 110-124 heme oxygenase 1 Homo sapiens 228-232 25813625-7 2015 Moreover, it was found that piperlongumine-induced Nrf2 activation, HO-1 expression and cancer cell apoptosis are not dependent on the generation of reactive oxygen species. piperlonguminine 28-42 heme oxygenase 1 Homo sapiens 68-72 25813625-8 2015 Instead, piperlongumine, which bears electrophilic alpha,beta-unsaturated carbonyl groups, appears to inactivate Kelch-like ECH-associated protein-1 (Keap1) through thiol modification, thereby activating the Nrf2/HO-1 pathway and subsequently upregulating HO-1 expression, which accounts for piperlongumine-induced apoptosis in cancer cells. piperlonguminine 9-23 heme oxygenase 1 Homo sapiens 213-217 25813625-8 2015 Instead, piperlongumine, which bears electrophilic alpha,beta-unsaturated carbonyl groups, appears to inactivate Kelch-like ECH-associated protein-1 (Keap1) through thiol modification, thereby activating the Nrf2/HO-1 pathway and subsequently upregulating HO-1 expression, which accounts for piperlongumine-induced apoptosis in cancer cells. piperlonguminine 9-23 heme oxygenase 1 Homo sapiens 256-260 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Heme 95-99 heme oxygenase 1 Homo sapiens 18-22 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Iron 117-121 heme oxygenase 1 Homo sapiens 0-16 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Iron 117-121 heme oxygenase 1 Homo sapiens 18-22 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Carbon Monoxide 123-138 heme oxygenase 1 Homo sapiens 0-16 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Carbon Monoxide 123-138 heme oxygenase 1 Homo sapiens 18-22 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Carbon Monoxide 140-142 heme oxygenase 1 Homo sapiens 0-16 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Carbon Monoxide 140-142 heme oxygenase 1 Homo sapiens 18-22 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Biliverdine 148-158 heme oxygenase 1 Homo sapiens 0-16 25885228-1 2015 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme catalyzing oxidative degradation of cellular heme to liberate free iron, carbon monoxide (CO) and biliverdin in mammalian cells. Biliverdine 148-158 heme oxygenase 1 Homo sapiens 18-22 25885228-2 2015 In addition to its primary role in heme catabolism, HO-1 exhibits anti-oxidative and anti-inflammatory functions via the actions of biliverdin and CO, respectively. Heme 35-39 heme oxygenase 1 Homo sapiens 52-56 25885228-2 2015 In addition to its primary role in heme catabolism, HO-1 exhibits anti-oxidative and anti-inflammatory functions via the actions of biliverdin and CO, respectively. Biliverdine 132-142 heme oxygenase 1 Homo sapiens 52-56 25885228-2 2015 In addition to its primary role in heme catabolism, HO-1 exhibits anti-oxidative and anti-inflammatory functions via the actions of biliverdin and CO, respectively. Carbon Monoxide 147-149 heme oxygenase 1 Homo sapiens 52-56 25686009-7 2015 Anthocyanin metabolites may therefore modulate vascular reactivity by inducing HO-1 and modulating NOX activity, resulting in reduced superoxide production and improved NO bioavailability. Anthocyanins 0-11 heme oxygenase 1 Homo sapiens 79-83 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Heme 77-81 heme oxygenase 1 Homo sapiens 0-16 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Heme 77-81 heme oxygenase 1 Homo sapiens 18-22 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Iron 90-94 heme oxygenase 1 Homo sapiens 18-22 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Iron 90-94 heme oxygenase 1 Homo sapiens 0-16 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Carbon Monoxide 96-111 heme oxygenase 1 Homo sapiens 0-16 25769104-5 2015 Glutathione-S-transferase and Heme Oxygenase 1 mRNA levels are induced with ADE PM and reduced by DF and NAC. Adenine 76-79 heme oxygenase 1 Homo sapiens 30-46 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Carbon Monoxide 96-111 heme oxygenase 1 Homo sapiens 18-22 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Biliverdine 116-126 heme oxygenase 1 Homo sapiens 0-16 25761244-1 2015 Heme oxygenase-1 (HO-1) is a 32 kDa protein which catalyzes the breakdown of heme to free iron, carbon monoxide and biliverdin. Biliverdine 116-126 heme oxygenase 1 Homo sapiens 18-22 25761244-3 2015 In "stressed" astroglia, HO-1 hyperactivity promotes mitochondrial iron sequestration and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure documented in Alzheimer disease, Parkinson disease and certain neurodevelopmental conditions. Iron 67-71 heme oxygenase 1 Homo sapiens 25-29 25761244-3 2015 In "stressed" astroglia, HO-1 hyperactivity promotes mitochondrial iron sequestration and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure documented in Alzheimer disease, Parkinson disease and certain neurodevelopmental conditions. Iron 152-156 heme oxygenase 1 Homo sapiens 25-29 25761244-4 2015 Glial HO-1 expression may also impact neuroplasticity and cell survival by modulating brain sterol metabolism and the proteasomal degradation of neurotoxic proteins. Sterols 92-98 heme oxygenase 1 Homo sapiens 6-10 25267105-12 2015 Besides, ES reduced TNF-alpha- and H2O2 -induced oxidative stress and in parallel induces anti-inflammatory haem oxygenase (HO)-1 expression. sesamin 9-11 heme oxygenase 1 Homo sapiens 108-129 25592228-7 2015 Transfection with ATM induced expression of HO-1 which was mediated by PKC-delta and NF-kappaB in H2O2-treated MOCK cells. Hydrogen Peroxide 98-102 heme oxygenase 1 Homo sapiens 44-48 26045831-0 2015 Effects of dexmedetomidine pretreatment on heme oxygenase-1 expression and oxidative stress during one-lung ventilation. Dexmedetomidine 11-26 heme oxygenase 1 Homo sapiens 43-59 26045831-1 2015 PURPOSE: This study aimed to explore effects of dexmedetomidine pretreatment on heme oxygenase-1 (HO-1) expression and oxidative stress during one-lung ventilation (OLV) in lung cancer patients. Dexmedetomidine 48-63 heme oxygenase 1 Homo sapiens 80-96 26045831-1 2015 PURPOSE: This study aimed to explore effects of dexmedetomidine pretreatment on heme oxygenase-1 (HO-1) expression and oxidative stress during one-lung ventilation (OLV) in lung cancer patients. Dexmedetomidine 48-63 heme oxygenase 1 Homo sapiens 98-102 26045831-6 2015 Superoxide dismutase (SOD) activity and the expression level of HO-1 were higher in Dexmedetomidine group than in Control (P<0.05). Dexmedetomidine 84-99 heme oxygenase 1 Homo sapiens 64-68 26045831-7 2015 CONCLUSIONS: Dexmedetomidine pretreatment could upregulated expression of HO-1 in lung tissue and reduce oxidative stress and inflammation during OLV. Dexmedetomidine 13-28 heme oxygenase 1 Homo sapiens 74-78 26045831-8 2015 Thus dexmedetomidine played a role in protecting lung injury by promoting HO-1 expression. Dexmedetomidine 5-20 heme oxygenase 1 Homo sapiens 74-78 25585641-10 2015 Upregulation of p16 and changing of p16-relative cell cycle protein was observed after silencing HO-1 in AZA treated SKM-1 cells. Azacitidine 105-108 heme oxygenase 1 Homo sapiens 97-101 25585641-12 2015 In vivo, silencing HO-1 inhibited SKM-1 cell growth induced by AZA in a NOD/SCID mouse model. Azacitidine 63-66 heme oxygenase 1 Homo sapiens 19-23 25585641-13 2015 Silencing HO-1 sensitized SKM-1 cells toward AZA, which may be attributed to the influence of HO-1 on AZA-induced p16 demethylation. Azacitidine 45-48 heme oxygenase 1 Homo sapiens 10-14 25585641-13 2015 Silencing HO-1 sensitized SKM-1 cells toward AZA, which may be attributed to the influence of HO-1 on AZA-induced p16 demethylation. Azacitidine 45-48 heme oxygenase 1 Homo sapiens 94-98 25585641-13 2015 Silencing HO-1 sensitized SKM-1 cells toward AZA, which may be attributed to the influence of HO-1 on AZA-induced p16 demethylation. Azacitidine 102-105 heme oxygenase 1 Homo sapiens 10-14 25585641-13 2015 Silencing HO-1 sensitized SKM-1 cells toward AZA, which may be attributed to the influence of HO-1 on AZA-induced p16 demethylation. Azacitidine 102-105 heme oxygenase 1 Homo sapiens 94-98 25585641-14 2015 HO-1 may be one of the targets that enhance the therapeutic effects of AZA on MDS malignant transformation inspiring new treatment methods for high-risk and very high-risk MDS patients in clinical practice. Azacitidine 71-74 heme oxygenase 1 Homo sapiens 0-4 25592228-8 2015 ZnPP, an HO-1 inhibitor, and transfection with HO-1 siRNA increased ROS levels and apoptosis, whereas hemin, an HO-1 activator, reduced ROS levels and apoptosis in H2O2-treated YZ5 cells. zinc protoporphyrin 0-4 heme oxygenase 1 Homo sapiens 9-13 25592228-8 2015 ZnPP, an HO-1 inhibitor, and transfection with HO-1 siRNA increased ROS levels and apoptosis, whereas hemin, an HO-1 activator, reduced ROS levels and apoptosis in H2O2-treated YZ5 cells. Reactive Oxygen Species 68-71 heme oxygenase 1 Homo sapiens 47-51 25592228-8 2015 ZnPP, an HO-1 inhibitor, and transfection with HO-1 siRNA increased ROS levels and apoptosis, whereas hemin, an HO-1 activator, reduced ROS levels and apoptosis in H2O2-treated YZ5 cells. Reactive Oxygen Species 68-71 heme oxygenase 1 Homo sapiens 47-51 25592228-8 2015 ZnPP, an HO-1 inhibitor, and transfection with HO-1 siRNA increased ROS levels and apoptosis, whereas hemin, an HO-1 activator, reduced ROS levels and apoptosis in H2O2-treated YZ5 cells. Hydrogen Peroxide 164-168 heme oxygenase 1 Homo sapiens 47-51 25592228-8 2015 ZnPP, an HO-1 inhibitor, and transfection with HO-1 siRNA increased ROS levels and apoptosis, whereas hemin, an HO-1 activator, reduced ROS levels and apoptosis in H2O2-treated YZ5 cells. Hydrogen Peroxide 164-168 heme oxygenase 1 Homo sapiens 47-51 25592228-9 2015 Rottlerin, a PKC-delta inhibitor, inhibited NF-kappaB activation and HO-1 expression in H2O2-treated YZ5 cells. rottlerin 0-9 heme oxygenase 1 Homo sapiens 69-73 25592228-9 2015 Rottlerin, a PKC-delta inhibitor, inhibited NF-kappaB activation and HO-1 expression in H2O2-treated YZ5 cells. Hydrogen Peroxide 88-92 heme oxygenase 1 Homo sapiens 69-73 25592228-11 2015 In addition, transfection with p65 siRNA increased ROS levels and DNA fragmentation, but decreased HO-1 protein levels in H2O2-treated YZ5 cells. Hydrogen Peroxide 122-126 heme oxygenase 1 Homo sapiens 99-103 25585641-0 2015 Silencing HO-1 sensitizes SKM-1 cells to apoptosis induced by low concentration 5-azacytidine through enhancing p16 demethylation. Azacitidine 80-93 heme oxygenase 1 Homo sapiens 10-14 25585641-2 2015 We found that HO-1 was resistant to 5-azacytidine (AZA) treatment of myelodysplastic syndrome (MDS), and explored further the relative mechanisms. Azacitidine 36-49 heme oxygenase 1 Homo sapiens 14-18 25585641-2 2015 We found that HO-1 was resistant to 5-azacytidine (AZA) treatment of myelodysplastic syndrome (MDS), and explored further the relative mechanisms. Azacitidine 51-54 heme oxygenase 1 Homo sapiens 14-18 25585641-7 2015 HO-1 overexpression was observed in SKM-1 cells after AZA treatment comparing to other cell lines. Azacitidine 54-57 heme oxygenase 1 Homo sapiens 0-4 25585641-9 2015 After HO-1 was silenced by lentivirus-mediated siRNA, the proliferation of SKM-1 cells was effectively inhibited by low concentration AZA, and the cell cycle was arrested in the G0/G1 phase. Azacitidine 134-137 heme oxygenase 1 Homo sapiens 6-10 25471815-0 2015 alpha-Tocopherol protects renal cells from nicotine- or oleic acid-provoked oxidative stress via inducing heme oxygenase-1. alpha-Tocopherol 0-16 heme oxygenase 1 Homo sapiens 106-122 25388553-2 2015 This study investigates the interaction between glucocorticoids or estrogens and GFs on the expression of heme oxygenase-1 (HO-1) and cyclin D1 in astrocyte cultures at 14 days treated for 48 or 60 hr with dexamethasone (DEX) or 48 hr with 17beta-estradiol (E2) alone or with GFs added only in the last 12 or 24 hr. Dexamethasone 206-219 heme oxygenase 1 Homo sapiens 106-122 25388553-2 2015 This study investigates the interaction between glucocorticoids or estrogens and GFs on the expression of heme oxygenase-1 (HO-1) and cyclin D1 in astrocyte cultures at 14 days treated for 48 or 60 hr with dexamethasone (DEX) or 48 hr with 17beta-estradiol (E2) alone or with GFs added only in the last 12 or 24 hr. Dexamethasone 221-224 heme oxygenase 1 Homo sapiens 106-122 25388553-2 2015 This study investigates the interaction between glucocorticoids or estrogens and GFs on the expression of heme oxygenase-1 (HO-1) and cyclin D1 in astrocyte cultures at 14 days treated for 48 or 60 hr with dexamethasone (DEX) or 48 hr with 17beta-estradiol (E2) alone or with GFs added only in the last 12 or 24 hr. Estradiol 240-256 heme oxygenase 1 Homo sapiens 106-122 25388553-3 2015 Twelve- or twenty-four-hour epidermal growth factor (EGF) treatment significantly enhanced HO-1 expression in astrocyte cultures pretreated for 48 hr with DEX. Dexamethasone 155-158 heme oxygenase 1 Homo sapiens 91-95 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). Vitamin E 143-152 heme oxygenase 1 Homo sapiens 248-264 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). Vitamin E 143-152 heme oxygenase 1 Homo sapiens 266-270 25471815-0 2015 alpha-Tocopherol protects renal cells from nicotine- or oleic acid-provoked oxidative stress via inducing heme oxygenase-1. Nicotine 43-51 heme oxygenase 1 Homo sapiens 106-122 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). alpha-Tocopherol 164-180 heme oxygenase 1 Homo sapiens 248-264 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). alpha-Tocopherol 164-180 heme oxygenase 1 Homo sapiens 266-270 25471815-0 2015 alpha-Tocopherol protects renal cells from nicotine- or oleic acid-provoked oxidative stress via inducing heme oxygenase-1. Oleic Acid 56-66 heme oxygenase 1 Homo sapiens 106-122 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). alpha-Tocopherol 182-185 heme oxygenase 1 Homo sapiens 248-264 25471815-1 2015 Smoking and obesity increases renal oxidative stress via nicotine (NIC) or free fatty acid such as oleic acid (OA) but decreases levels of the vitamin E-derivative alpha-tocopherol (TOC), which has shown to stimulate the antioxidant system such as heme oxygenase-1 (HO-1). alpha-Tocopherol 182-185 heme oxygenase 1 Homo sapiens 266-270 25659595-4 2015 In addition, increased heme oxygenase-1 (HMOX1) expression and non-toxic generation of superoxide were detected in the first 4 h. Regarding the effects on the UVB-induced toxicity, although the level of cyclobutane pyrimidine dimers in the keratinocytes pre-treated with zinc for 24 h was reduced 3 h after UVB irradiation, significantly enhanced superoxide generation was observed 10 h after UVB exposure in the zinc pre-exposed cells. cyclobutane pyrimidine 203-225 heme oxygenase 1 Homo sapiens 41-46 25471815-3 2015 NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Reactive Oxygen Species 35-58 heme oxygenase 1 Homo sapiens 168-172 25471815-3 2015 NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Reactive Oxygen Species 35-58 heme oxygenase 1 Homo sapiens 213-217 25471815-3 2015 NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Reactive Oxygen Species 60-63 heme oxygenase 1 Homo sapiens 168-172 25471815-3 2015 NIC- or OA-dependent production of reactive oxygen species (ROS) was determined in the presence or absence of various pharmacologic or genetic inhibitors that modulate HO-1 activation and enhancer elements in the HO-1 promoter such as the antioxidant response element (ARE) and the cAMP-response element (CRE) in renal proximal tubule cells (NRK52E). Reactive Oxygen Species 60-63 heme oxygenase 1 Homo sapiens 213-217 25471815-5 2015 We found that pre- or posttreatment with TOC attenuated NIC- or OA-dependent ROS production that required HO-1 activation. alpha-Tocopherol 41-44 heme oxygenase 1 Homo sapiens 106-110 25471815-5 2015 We found that pre- or posttreatment with TOC attenuated NIC- or OA-dependent ROS production that required HO-1 activation. Reactive Oxygen Species 77-80 heme oxygenase 1 Homo sapiens 106-110 25471815-8 2015 Hence, vitamin E supplementation-via induction of the cytoprotective HO-1-may help to reduce renal oxidative stress imposed by smoking or obesity. Vitamin E 7-16 heme oxygenase 1 Homo sapiens 69-73 26269017-1 2015 OBJECTIVE: To investigate the relationship between (GT)n polymorphism and esophageal cancer by analyzing the connection between microsatellite polymorphisms in the promoter of heme oxygenase-1 and the clinicopathological characteristics of esophageal squamous cell carcinoma (ESCC) in Han chinese population. Nitrogen 15-16 heme oxygenase 1 Homo sapiens 176-192 25759595-5 2015 We found that PEMF treatment was followed by changes in the relative amount of messenger (m)RNAs encoding enzymes involved in heme catabolism and removal of reactive oxygen species, including an increase in heme oxygenase 1 and superoxide dismutase 3 mRNAs, in all cell types examined 2 hours after PEMF treatment. pemf 14-18 heme oxygenase 1 Homo sapiens 207-223 26269017-5 2015 For in vitro experiments, the transient-transfection assay was performed to explore the correlation between different lengths of (GT)n repeats and promoter activity by assessing the promoter activities of HO-1 gene in cultured Ecal09 cells treated with H2O2 by analysis of cariance. Hydrogen Peroxide 253-257 heme oxygenase 1 Homo sapiens 205-209 26269017-27 2015 In H2O treated control group, compared to (GT)26 and (GT)36, the basal promoter activities of HO-1 gene carrying (GT)16 repeat increased (F =41. Water 3-6 heme oxygenase 1 Homo sapiens 94-98 25196646-0 2015 Modulation of heme oxygenase-1 by metalloporphyrins increases anti-viral T cell responses. Metalloporphyrins 34-51 heme oxygenase 1 Homo sapiens 14-30 25541286-4 2015 Shikonin (0-1 muM) concentration-dependently induced heme oxygenase-1, glutamate cysteine ligase modifier subunit, catalase, superoxide dismutase 1, glutathione peroxidase 1, and glutathione reductase protein and mRNA expression and glutathione content via activation of the phosphatidylinositol 3-kinase (PI3K)/Akt/Nrf2 signaling pathway. shikonin 0-8 heme oxygenase 1 Homo sapiens 53-69 25557764-8 2015 Thus an increase in Sirt4, Nrf2, Tfam, UCP1, eNOS, HO1 and STAT3 genes could profoundly affect mitochondrial function, cholesterol homeostasis, and fatty acid oxidation, suggesting that ASA could be beneficial beyond simply its ability to inhibit cyclooxygenase. Cholesterol 119-130 heme oxygenase 1 Homo sapiens 51-54 25557764-8 2015 Thus an increase in Sirt4, Nrf2, Tfam, UCP1, eNOS, HO1 and STAT3 genes could profoundly affect mitochondrial function, cholesterol homeostasis, and fatty acid oxidation, suggesting that ASA could be beneficial beyond simply its ability to inhibit cyclooxygenase. Fatty Acids 148-158 heme oxygenase 1 Homo sapiens 51-54 25557764-8 2015 Thus an increase in Sirt4, Nrf2, Tfam, UCP1, eNOS, HO1 and STAT3 genes could profoundly affect mitochondrial function, cholesterol homeostasis, and fatty acid oxidation, suggesting that ASA could be beneficial beyond simply its ability to inhibit cyclooxygenase. Aspirin 186-189 heme oxygenase 1 Homo sapiens 51-54 25484350-5 2015 Treatment with tBHQ induced mRNA expression of the Nrf2 target genes HMOX-1, GCLC, and NQO1, and also increased NRF2 mRNA expression, albeit to a lesser extent than the other target genes. 2-tert-butylhydroquinone 15-19 heme oxygenase 1 Homo sapiens 69-75 25483558-10 2015 Whereas, treatment with zinc protoporphyrin IX (a HO-1 inhibitor) into the IPC+ischemia-operated groups did not preserve the IPC-mediated increase of HO-1 and lost beneficial effects of IPC by inhibiting ischemia-induced DNA damage and lipid peroxidation. protoporphyrin IX 29-46 heme oxygenase 1 Homo sapiens 50-54 24744106-0 2015 Heme oxygenase-1 regulates cell proliferation via carbon monoxide-mediated inhibition of T-type Ca2+ channels. Carbon Monoxide 50-65 heme oxygenase 1 Homo sapiens 0-16 25499849-4 2015 Microarray analysis of HaCaT cells revealed that 10-EZ-HODE and 12-ZE-HODE induced cellular antioxidant genes that are responsive to nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), such as heme oxygenase-1 and glutathione synthesis enzymes. 10-ez-hode 49-59 heme oxygenase 1 Homo sapiens 197-213 25499849-4 2015 Microarray analysis of HaCaT cells revealed that 10-EZ-HODE and 12-ZE-HODE induced cellular antioxidant genes that are responsive to nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), such as heme oxygenase-1 and glutathione synthesis enzymes. 12-ze-hode 64-74 heme oxygenase 1 Homo sapiens 197-213 25499849-4 2015 Microarray analysis of HaCaT cells revealed that 10-EZ-HODE and 12-ZE-HODE induced cellular antioxidant genes that are responsive to nuclear factor-erythroid 2 p45-related factor 2 (Nrf2), such as heme oxygenase-1 and glutathione synthesis enzymes. Glutathione 218-229 heme oxygenase 1 Homo sapiens 197-213 25393640-10 2015 CONCLUSIONS: In a 2-months prospective pilot study, the addition of liraglutide to metformin resulted in improvement in oxidative stress as well as plasma ghrelin and HO-1 concentrations in patients with T2DM. Metformin 83-92 heme oxygenase 1 Homo sapiens 167-171 25611090-5 2015 Furthermore, the cell-intrinsic and -extrinsic factors (namely, Spi-C, IRF8/4, heme oxygenase-1, and M-CSF) that regulate the development and survival of RPMs have been identified. rpms 154-158 heme oxygenase 1 Homo sapiens 71-95 24744106-4 2015 Proliferation and [Ca(2+)]i levels were reduced to levels seen in non-transfected cells either by induction of HO-1 or exposure of cells to the HO-1 product, carbon monoxide (CO) (applied as the CO releasing molecule, CORM-3). Carbon Monoxide 158-173 heme oxygenase 1 Homo sapiens 144-148 25593219-1 2015 Sulforaphane (SFN), one of the most important isothiocyanates in the human diet, is known to have chemo-preventive and antioxidant activities in different tissues via activation of nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 and NAD(P)H quinone oxidoreductase 1. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 292-345 25988128-8 2014 EGCG improved efficacy of cisplatin treatment in HeLa cells by regulating NFkappaB p65, COX-2, p-Akt, and p-mTOR pathways, whereas it increased the expression levels of Nrf2/HO-1 in combined therapy. epigallocatechin gallate 0-4 heme oxygenase 1 Homo sapiens 174-178 25988128-8 2014 EGCG improved efficacy of cisplatin treatment in HeLa cells by regulating NFkappaB p65, COX-2, p-Akt, and p-mTOR pathways, whereas it increased the expression levels of Nrf2/HO-1 in combined therapy. Cisplatin 26-35 heme oxygenase 1 Homo sapiens 174-178 25593219-1 2015 Sulforaphane (SFN), one of the most important isothiocyanates in the human diet, is known to have chemo-preventive and antioxidant activities in different tissues via activation of nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 and NAD(P)H quinone oxidoreductase 1. sulforaphane 14-17 heme oxygenase 1 Homo sapiens 292-345 26303493-3 2015 HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. Heme 14-18 heme oxygenase 1 Homo sapiens 0-4 25574604-2 2015 We wanted to evaluate whether a functional polymorphism in the HMOX1 gene encoding heme oxygenase modifies risk of CRC or interacts with diet or lifestyle factors because this would identify heme or heme iron as a risk factor of CRC. Heme 83-87 heme oxygenase 1 Homo sapiens 63-68 25574604-2 2015 We wanted to evaluate whether a functional polymorphism in the HMOX1 gene encoding heme oxygenase modifies risk of CRC or interacts with diet or lifestyle factors because this would identify heme or heme iron as a risk factor of CRC. Heme 191-195 heme oxygenase 1 Homo sapiens 63-68 25574604-2 2015 We wanted to evaluate whether a functional polymorphism in the HMOX1 gene encoding heme oxygenase modifies risk of CRC or interacts with diet or lifestyle factors because this would identify heme or heme iron as a risk factor of CRC. Iron 204-208 heme oxygenase 1 Homo sapiens 63-68 25610397-1 2014 The heme-degrading enzyme heme oxygenase-1 (HO-1) has cytoprotective, antioxidant, and anti-inflammatory properties. Heme 4-8 heme oxygenase 1 Homo sapiens 26-42 25610397-1 2014 The heme-degrading enzyme heme oxygenase-1 (HO-1) has cytoprotective, antioxidant, and anti-inflammatory properties. Heme 4-8 heme oxygenase 1 Homo sapiens 44-48 26303493-3 2015 HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. Carbon Monoxide 31-46 heme oxygenase 1 Homo sapiens 0-4 26303493-3 2015 HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. Carbon Monoxide 48-50 heme oxygenase 1 Homo sapiens 0-4 26303493-3 2015 HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. ammonium ferrous sulfate 63-69 heme oxygenase 1 Homo sapiens 0-4 26303493-3 2015 HO-1 degrades heme to generate carbon monoxide (CO) along with Fe(2+) and biliverdin. Biliverdine 74-84 heme oxygenase 1 Homo sapiens 0-4 26295053-2 2015 We hypothesized that glutathione S-transferase (GST M1, T1, and P1), reactive oxygen species (ROS) related metabolic genes (NQO1, EPHX1, and HO-1), and DNA repair genes (XRCC1, XPD, hOGG1, and ATM) together may play a role in arsenic-induced skin carcinogenesis. Reactive Oxygen Species 94-97 heme oxygenase 1 Homo sapiens 141-145 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Heme 55-59 heme oxygenase 1 Homo sapiens 0-16 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Heme 55-59 heme oxygenase 1 Homo sapiens 18-22 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Bilirubin 65-74 heme oxygenase 1 Homo sapiens 0-16 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Bilirubin 65-74 heme oxygenase 1 Homo sapiens 18-22 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Carbon Monoxide 76-91 heme oxygenase 1 Homo sapiens 0-16 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Carbon Monoxide 76-91 heme oxygenase 1 Homo sapiens 18-22 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Iron 97-101 heme oxygenase 1 Homo sapiens 0-16 25744452-1 2015 Heme oxygenase-1 (HO-1) catabolizes the degradation of heme into bilirubin, carbon monoxide, and iron ions. Iron 97-101 heme oxygenase 1 Homo sapiens 18-22 25744452-3 2015 HO-1 is induced by its substrate heme and environmental factors including oxidative and heat stresses. Heme 33-37 heme oxygenase 1 Homo sapiens 0-4 25744452-6 2015 Based on a comparison of the results obtained with these cells, we found that nitric oxide (NO) was closely linked to the induction of HO-1. Nitric Oxide 78-90 heme oxygenase 1 Homo sapiens 135-139 25744452-9 2015 N-Nitro-L-arginine methyl ester, an NO synthase inhibitor, reduced both HO-1 expression and NO production in LPS+IFN-gamma-treated JA-4 cells. n-nitro-l-arginine methyl ester 0-31 heme oxygenase 1 Homo sapiens 72-76 25744452-12 2015 These results suggest that, among ROS, NO plays an important role in HO-1 induction in activated macrophages treated with LPS+IFN-gamma. Reactive Oxygen Species 34-37 heme oxygenase 1 Homo sapiens 69-73 25661467-0 2015 High glucose promotes tumor invasion and increases metastasis-associated protein expression in human lung epithelial cells by upregulating heme oxygenase-1 via reactive oxygen species or the TGF-beta1/PI3K/Akt signaling pathway. Glucose 5-12 heme oxygenase 1 Homo sapiens 139-155 25661467-0 2015 High glucose promotes tumor invasion and increases metastasis-associated protein expression in human lung epithelial cells by upregulating heme oxygenase-1 via reactive oxygen species or the TGF-beta1/PI3K/Akt signaling pathway. Reactive Oxygen Species 160-183 heme oxygenase 1 Homo sapiens 139-155 25661467-1 2015 BACKGROUND: Growing evidence indicates that heme oxygenase-1 (HO-1) is up-regulated in malignancies and subsequently alters tumor aggressiveness and various cancer-related factors, such as high glucose (HG) levels. Glucose 194-201 heme oxygenase 1 Homo sapiens 44-60 25661467-1 2015 BACKGROUND: Growing evidence indicates that heme oxygenase-1 (HO-1) is up-regulated in malignancies and subsequently alters tumor aggressiveness and various cancer-related factors, such as high glucose (HG) levels. Glucose 194-201 heme oxygenase 1 Homo sapiens 62-66 25661467-2 2015 HO-1 expression can be induced when glucose concentrations are above 25 mM; however, the role of HO-1 in lung cancer patients with diabetes remains unknown. Glucose 36-43 heme oxygenase 1 Homo sapiens 0-4 25661467-7 2015 RESULTS: HG treatment of A549 cells induced an increase in HO-1 expression, which was mediated by the HG-induced generation of reactive oxygen species (ROS) and transforming growth factor-beta1 (TGF-beta1) in a concentration- and time-dependent manner. Reactive Oxygen Species 127-150 heme oxygenase 1 Homo sapiens 59-63 25661467-7 2015 RESULTS: HG treatment of A549 cells induced an increase in HO-1 expression, which was mediated by the HG-induced generation of reactive oxygen species (ROS) and transforming growth factor-beta1 (TGF-beta1) in a concentration- and time-dependent manner. Reactive Oxygen Species 152-155 heme oxygenase 1 Homo sapiens 59-63 26159150-0 2015 Induction of Heme Oxygenase-1 by Sodium 9-Hydroxyltanshinone IIA Sulfonate Derivative Contributes to Inhibit LPS-Mediated Inflammatory Response in Macrophages. sodium 9-hydroxyltanshinone iia sulfonate 33-74 heme oxygenase 1 Homo sapiens 13-29 26159150-6 2015 In addition, ZY-1A4 concentration- and time-dependently induced HO-1 expression associated with degradation of Kelch-like ECH-associated protein 1 (Keap1) and nuclear translocation of Nrf2, while the effect of ZY-1A4 was abolished by a phosphoinositide 3-kinase (PI3K) inhibitor LY294002. zy-1a4 13-19 heme oxygenase 1 Homo sapiens 64-68 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. zy-1a4 161-167 heme oxygenase 1 Homo sapiens 46-50 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. zy-1a4 161-167 heme oxygenase 1 Homo sapiens 192-196 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. Carbon Monoxide 209-224 heme oxygenase 1 Homo sapiens 46-50 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. Carbon Monoxide 209-224 heme oxygenase 1 Homo sapiens 192-196 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. Bilirubin 229-238 heme oxygenase 1 Homo sapiens 46-50 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. Bilirubin 229-238 heme oxygenase 1 Homo sapiens 192-196 26159150-8 2015 Furthermore, the inhibitory effect of ZY-1A4 on LPS-induced iNOS expression and NO release was abolished by HO-1 siRNA or LY294002. zy-1a4 38-44 heme oxygenase 1 Homo sapiens 108-112 26159150-6 2015 In addition, ZY-1A4 concentration- and time-dependently induced HO-1 expression associated with degradation of Kelch-like ECH-associated protein 1 (Keap1) and nuclear translocation of Nrf2, while the effect of ZY-1A4 was abolished by a phosphoinositide 3-kinase (PI3K) inhibitor LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 279-287 heme oxygenase 1 Homo sapiens 64-68 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. zinc protoporphyrin 56-78 heme oxygenase 1 Homo sapiens 46-50 26159150-7 2015 Intriguingly, pharmacological inactivation of HO-1 with zinc protoporphyrin IX reversed anti-inflammatory effect of ZY- 1A4, but the anti-inflammatory effect of ZY-1A4 was largely mimicked by HO-1 by-products carbon monoxide and bilirubin. zy- 1a4 116-123 heme oxygenase 1 Homo sapiens 46-50 26151294-0 2015 Edaravone protects rats and human pulmonary alveolar epithelial cells against hyperoxia injury: heme oxygenase-1 and PI3K/Akt pathway may be involved. Edaravone 0-9 heme oxygenase 1 Homo sapiens 96-112 25874340-0 2015 Novel imidazole derivatives as heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2) inhibitors and their cytotoxic activity in human-derived cancer cell lines. imidazole 6-15 heme oxygenase 1 Homo sapiens 31-47 25874340-0 2015 Novel imidazole derivatives as heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2) inhibitors and their cytotoxic activity in human-derived cancer cell lines. imidazole 6-15 heme oxygenase 1 Homo sapiens 49-53 25874340-2 2015 In this work, novel imidazole derivatives were designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). imidazole 20-29 heme oxygenase 1 Homo sapiens 89-105 25874340-2 2015 In this work, novel imidazole derivatives were designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). imidazole 20-29 heme oxygenase 1 Homo sapiens 107-111 26495956-9 2015 qRT-PCR analysis confirmed significant upregulation of SULF2, IGF2, and HMOX1 in graft regions with PFA. pfa 100-103 heme oxygenase 1 Homo sapiens 72-77 25527214-1 2015 Pegylated zinc protoporphyrin (PEG-ZnPP) is a water-soluble inhibitor of heme oxygenase-1. zinc protoporphyrin 10-29 heme oxygenase 1 Homo sapiens 73-89 25527214-1 2015 Pegylated zinc protoporphyrin (PEG-ZnPP) is a water-soluble inhibitor of heme oxygenase-1. pegylated zinc protoporphyrin 31-39 heme oxygenase 1 Homo sapiens 73-89 25527214-1 2015 Pegylated zinc protoporphyrin (PEG-ZnPP) is a water-soluble inhibitor of heme oxygenase-1. Water 46-51 heme oxygenase 1 Homo sapiens 73-89 26151294-9 2015 CONCLUSIONS: This study indicates that edaravone protects rats and human pulmonary alveolar epithelial cells against hyperoxia-induced injury and the antioxidant effect may be related to upregulation of HO-1, which is regulated by PI3K/Akt pathway. Edaravone 39-48 heme oxygenase 1 Homo sapiens 203-207 25084759-5 2015 The neuroprotective effect of agmatine was abolished by yohimbine (alpha2-adrenoceptor antagonist), ketanserin (5-HT2A receptor antagonist), LY294002 (PI3K inhibitor), PD98059 (MEK1/2 inhibitor), SnPP (HO-1 inhibitor), and cycloheximide (protein synthesis inhibitor). Agmatine 30-38 heme oxygenase 1 Homo sapiens 202-206 25463280-1 2015 Heme oxygenase (HO)-1 is the inducible isoform of the heme-degrading enzyme HO, which is upregulated by multiple stress stimuli. Heme 54-58 heme oxygenase 1 Homo sapiens 0-21 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. Hemin 48-53 heme oxygenase 1 Homo sapiens 13-29 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. Hemin 48-53 heme oxygenase 1 Homo sapiens 31-36 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. Hemin 48-53 heme oxygenase 1 Homo sapiens 37-41 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. chelerythrine 67-80 heme oxygenase 1 Homo sapiens 13-29 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. chelerythrine 67-80 heme oxygenase 1 Homo sapiens 31-36 25185584-8 2015 Induction of heme-oxygenase-1 (HMOX1/HO-1) with hemin also blocked chelerythrine-induced cell death. chelerythrine 67-80 heme oxygenase 1 Homo sapiens 37-41 25553444-3 2015 Exposure to isoflurane decreased miR-155 and upregulated HIF-1 alpha and HO-1 mRNA and protein. Isoflurane 12-22 heme oxygenase 1 Homo sapiens 73-77 25553444-6 2015 These results suggest that isoflurane posttreatment hr alleviates LPS-induced ALI and cell injury by triggering miR-155-HIF-1 alpha pathway, leading to upregulation of HO-1. Isoflurane 27-37 heme oxygenase 1 Homo sapiens 168-172 25084759-6 2015 Agmatine increased Akt and ERK phosphorylation and induced the transcription factor Nrf2 and the proteins HO-1 and GCLc; induction of these proteins was prevented by yohimbine, ketanserin, LY294002, and PD98059. Agmatine 0-8 heme oxygenase 1 Homo sapiens 106-119 25084759-6 2015 Agmatine increased Akt and ERK phosphorylation and induced the transcription factor Nrf2 and the proteins HO-1 and GCLc; induction of these proteins was prevented by yohimbine, ketanserin, LY294002, and PD98059. Ketanserin 177-187 heme oxygenase 1 Homo sapiens 106-119 25084759-7 2015 In conclusion, agmatine affords neuroprotection against corticosterone effects by a mechanism that implicates Nrf2 induction via alpha2-adrenergic and 5-HT2A receptors, Akt and ERK pathways, and HO-1 and GCLc expression. Agmatine 15-23 heme oxygenase 1 Homo sapiens 195-199 25084759-7 2015 In conclusion, agmatine affords neuroprotection against corticosterone effects by a mechanism that implicates Nrf2 induction via alpha2-adrenergic and 5-HT2A receptors, Akt and ERK pathways, and HO-1 and GCLc expression. Corticosterone 56-70 heme oxygenase 1 Homo sapiens 195-199 25876966-0 2015 [The protein expression of heme oxygenase-1 and platelet endothelial cell adhesion molecules-1 in human coronary artery endothelial cell induced by zinc oxide nanoparticle]. Zinc Oxide 148-158 heme oxygenase 1 Homo sapiens 27-43 25405261-4 2015 There were major differences between the types of NMs; exposure to ZnO and Ag resulted in cytotoxicity and increased gene expression levels of HMOX1 and IL8. Zinc Oxide 67-70 heme oxygenase 1 Homo sapiens 143-148 26161238-3 2015 Such protective phenomenon is reported to be implicated in decreasing the formation and reaction of reactive oxygen species and pro-nflammatory cytokines, as well as the mediation of a variety of intracellular signaling pathways, including the nitric oxide synthase, nicotinamide adenine dinucleotide phosphate oxidase, deacetylase sirtuin 1, mitogen-activated protein kinase, peroxisome proliferator-activated receptor-gamma coactivator 1 alpha, hemeoxygenase-1, and estrogen receptor-related pathways. Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 447-485 25926128-6 2015 In addition to a higher induction of the key cytoprotective gene heme oxygenase-1 (38.9-fold increase for CDDO-Im and 26.5-fold increase for CDDO-Me), CDDO-Im also induced greater expression of heat shock proteins (5.5-fold increase compared to 2.8-fold for CDDO-Me). 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 106-113 heme oxygenase 1 Homo sapiens 65-81 25926128-6 2015 In addition to a higher induction of the key cytoprotective gene heme oxygenase-1 (38.9-fold increase for CDDO-Im and 26.5-fold increase for CDDO-Me), CDDO-Im also induced greater expression of heat shock proteins (5.5-fold increase compared to 2.8-fold for CDDO-Me). bardoxolone methyl 141-148 heme oxygenase 1 Homo sapiens 65-81 25926128-6 2015 In addition to a higher induction of the key cytoprotective gene heme oxygenase-1 (38.9-fold increase for CDDO-Im and 26.5-fold increase for CDDO-Me), CDDO-Im also induced greater expression of heat shock proteins (5.5-fold increase compared to 2.8-fold for CDDO-Me). 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 151-158 heme oxygenase 1 Homo sapiens 65-81 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Heme 47-51 heme oxygenase 1 Homo sapiens 0-16 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Heme 47-51 heme oxygenase 1 Homo sapiens 18-22 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 0-16 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 18-22 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 0-16 26689007-1 2015 Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 18-22 25876966-1 2015 OBJECTIVE: To explore the protein expression of heme oxygenase-1 (HO-1) and platelet endothelial cell adhesion molecules-1 (PECAM-1) in human coronary artery endothelial cells induced with Zinc Oxide Nanoparticle (ZnO-NPs). Zinc Oxide 189-199 heme oxygenase 1 Homo sapiens 66-70 25876966-1 2015 OBJECTIVE: To explore the protein expression of heme oxygenase-1 (HO-1) and platelet endothelial cell adhesion molecules-1 (PECAM-1) in human coronary artery endothelial cells induced with Zinc Oxide Nanoparticle (ZnO-NPs). Zinc Oxide 214-217 heme oxygenase 1 Homo sapiens 48-64 25876966-4 2015 Protein levels of HO-1 (ng/L) in each group were 0.041+-0.011, 0.512+-0.076, 0.906+-0.059, 1.062+-0.089 respectively after the stimulation of different concentrations of ZnO-NPs (0, 10, 20, 40microg/ml). Zinc Oxide 170-173 heme oxygenase 1 Homo sapiens 18-22 25876966-1 2015 OBJECTIVE: To explore the protein expression of heme oxygenase-1 (HO-1) and platelet endothelial cell adhesion molecules-1 (PECAM-1) in human coronary artery endothelial cells induced with Zinc Oxide Nanoparticle (ZnO-NPs). Zinc Oxide 189-199 heme oxygenase 1 Homo sapiens 48-64 25404134-3 2014 Here, we report that multiple nuclear-enriched eRNAs are transcribed from the regions adjacent to two human HO-1 enhancers (i.e. the distal E2 and proximal E1 enhancers), and some of these eRNAs are induced by the oxidative stress-causing reagent diethyl maleate (DEM). diethyl maleate 247-262 heme oxygenase 1 Homo sapiens 108-112 25404134-4 2014 We demonstrated that the expression of one forward direction (5" to 3") eRNA transcribed from the human HO-1 E2 enhancer region (named human HO-1enhancer RNA E2-3; hereafter called eRNA E2-3) was induced by DEM in an NRF2-dependent manner in HeLa cells. diethyl maleate 207-210 heme oxygenase 1 Homo sapiens 104-108 25236743-5 2014 Early DNFB induction of ATF4 positively modulates autophagy-related genes MAP1LC3B and ATG3 and stabilizes the transcription factor Nrf2, causing a strong induction of the HMOX1-detoxifying gene. Dinitrofluorobenzene 6-10 heme oxygenase 1 Homo sapiens 172-177 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Heme 80-84 heme oxygenase 1 Homo sapiens 0-16 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Heme 80-84 heme oxygenase 1 Homo sapiens 18-22 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 0-16 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 18-22 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Iron 132-136 heme oxygenase 1 Homo sapiens 0-16 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Iron 132-136 heme oxygenase 1 Homo sapiens 18-22 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Biliverdine 142-152 heme oxygenase 1 Homo sapiens 0-16 25241054-1 2014 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting enzymatic step of heme degradation and produces carbon monoxide, free iron, and biliverdin. Biliverdine 142-152 heme oxygenase 1 Homo sapiens 18-22 25058038-1 2014 Carbon monoxide derived from the catalytic action of heme oxygenase-1 or carbon monoxide-releasing molecules (CORMs) has been found to potentially be an anticoagulant or procoagulant agent. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 53-69 25058038-1 2014 Carbon monoxide derived from the catalytic action of heme oxygenase-1 or carbon monoxide-releasing molecules (CORMs) has been found to potentially be an anticoagulant or procoagulant agent. Carbon Monoxide 73-88 heme oxygenase 1 Homo sapiens 53-69 25501830-0 2014 Heme oxygenase-1 protects against Alzheimer"s amyloid-beta(1-42)-induced toxicity via carbon monoxide production. Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Heme 94-98 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Heme 94-98 heme oxygenase 1 Homo sapiens 18-22 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Biliverdine 102-112 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Biliverdine 102-112 heme oxygenase 1 Homo sapiens 18-22 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). ammonium ferrous sulfate 114-118 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). ammonium ferrous sulfate 114-118 heme oxygenase 1 Homo sapiens 18-22 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Carbon Monoxide 123-138 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Carbon Monoxide 123-138 heme oxygenase 1 Homo sapiens 18-22 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Carbon Monoxide 140-142 heme oxygenase 1 Homo sapiens 0-16 25501830-1 2014 Heme oxygenase-1 (HO-1), an inducible enzyme up-regulated in Alzheimer"s disease, catabolises heme to biliverdin, Fe2+ and carbon monoxide (CO). Carbon Monoxide 140-142 heme oxygenase 1 Homo sapiens 18-22 25111043-4 2014 Induction of the glial HMOX1 gene may lead to pathological brain iron deposition, intracellular oxidative damage, and bioenergetic failure in AD and other human CNS disorders such as PD and MS. Iron 65-69 heme oxygenase 1 Homo sapiens 23-28 25231232-0 2014 Potential crosstalk of Ca2+-ROS-dependent mechanism involved in apoptosis of Kasumi-1 cells mediated by heme oxygenase-1 small interfering RNA. ca2+-ros 23-31 heme oxygenase 1 Homo sapiens 104-120 25063314-3 2014 HO-1 expression was assessed in 215 formalin-fixed bladder cancer specimens by immunohistochemistry. Formaldehyde 36-44 heme oxygenase 1 Homo sapiens 0-4 25227700-3 2014 To enhance intrainfarct graft viability, human embryonic stem cell-derived cardiomyocytes (hESC-CMs) were pretreated before implantation with cobalt protoporphyrin (CoPP), a pharmacologic inducer of cytoprotective heme oxygenase-1. cobaltiprotoporphyrin 142-163 heme oxygenase 1 Homo sapiens 214-230 25437431-5 2014 Additional tests showed that BQ is also capable of inducing expression of NADPH oxidase1 (NOX1), and several other antioxidant enzymes or drug-metabolizing enzymes, including heme oxygenase 1 (HMOX1), superoxide dismutase (SOD), catalase and NAD(P)H dehydrogenase quinone 1 (NQO1). quinone 29-31 heme oxygenase 1 Homo sapiens 175-191 25437431-5 2014 Additional tests showed that BQ is also capable of inducing expression of NADPH oxidase1 (NOX1), and several other antioxidant enzymes or drug-metabolizing enzymes, including heme oxygenase 1 (HMOX1), superoxide dismutase (SOD), catalase and NAD(P)H dehydrogenase quinone 1 (NQO1). quinone 29-31 heme oxygenase 1 Homo sapiens 193-198 25262300-1 2014 OBJECTIVE: To explore the effects of variants in Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) and Heme Oxygenase-1 (HMOX1) on daily physiological bilirubin levels and bilirubin changes during the first week after birth in Chinese newborns. Bilirubin 159-168 heme oxygenase 1 Homo sapiens 111-127 25262300-1 2014 OBJECTIVE: To explore the effects of variants in Uridine Diphosphate Glucuronosyl Transferase 1A1 (UGT1A1) and Heme Oxygenase-1 (HMOX1) on daily physiological bilirubin levels and bilirubin changes during the first week after birth in Chinese newborns. Bilirubin 159-168 heme oxygenase 1 Homo sapiens 129-134 25262300-2 2014 Both UGT1A1 and HMOX1 code rate-limiting enzymes in the bilirubin metabolism pathway. Bilirubin 56-65 heme oxygenase 1 Homo sapiens 16-21 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Ribavirin 0-9 heme oxygenase 1 Homo sapiens 109-125 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Ribavirin 0-9 heme oxygenase 1 Homo sapiens 127-132 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Heme 64-68 heme oxygenase 1 Homo sapiens 109-125 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Heme 64-68 heme oxygenase 1 Homo sapiens 127-132 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Carbon Monoxide 137-152 heme oxygenase 1 Homo sapiens 109-125 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Carbon Monoxide 137-152 heme oxygenase 1 Homo sapiens 127-132 25473172-9 2014 Ribavirin-induced hemolysis floods the hepatocytes and KCs with heme, which is metabolized and detoxified by heme oxygenase-1 (HMOX1) to carbon monoxide (CO), biliverdin and free iron (which induces ferritin). Iron 179-183 heme oxygenase 1 Homo sapiens 127-132 25450364-12 2014 GABA treatment significantly impeded the ALA-mediated increase in the number of hemoglobinized cells as well as the induction of HBG, HBA, and HMOX1. gamma-Aminobutyric Acid 0-4 heme oxygenase 1 Homo sapiens 143-148 25230250-6 2014 We also observed inductions (>2 fold) of genes involved in eicosanoid biosynthesis (Pla2g4a, Pla2g4b, Ptgs2, Ptgis, Alox12, Alox15), heme metabolism (Hpx, Hmox1, Prdx1) and iron homeostasis (Hamp, Tf). Eicosanoids 62-72 heme oxygenase 1 Homo sapiens 158-163 25450364-13 2014 Finally, small-interfering RNA-mediated knockdown of ALAS2 in HiDEP cells considerably decreased the expression of HBA, HBG, and HMOX1, and these expression levels were rescued with ALA treatment. 5-amino levulinic acid 53-56 heme oxygenase 1 Homo sapiens 129-134 24382006-3 2014 This may be, in part, explained by the high lung content of heme oxygenase-1 (HO-1), the rate-limiting enzyme in the degradation of heme and an important stress protein. Heme 60-64 heme oxygenase 1 Homo sapiens 78-82 25224540-2 2014 The products of HO-1 enzymatic activity, biliverdin and carbon monoxide (CO), actively contribute to this protection, suggesting that exploitation of these cellular systems may offer new therapeutic avenues in a variety of diseases. Biliverdine 41-51 heme oxygenase 1 Homo sapiens 16-20 25224540-2 2014 The products of HO-1 enzymatic activity, biliverdin and carbon monoxide (CO), actively contribute to this protection, suggesting that exploitation of these cellular systems may offer new therapeutic avenues in a variety of diseases. Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 16-20 25224540-2 2014 The products of HO-1 enzymatic activity, biliverdin and carbon monoxide (CO), actively contribute to this protection, suggesting that exploitation of these cellular systems may offer new therapeutic avenues in a variety of diseases. Carbon Monoxide 73-75 heme oxygenase 1 Homo sapiens 16-20 25033992-6 2014 Thereafter, we introduce some information covering melatonin-related signaling pathways, including heme oxygenase-1 (HO-1), toll-like receptor (TLR), c-Jun N-terminal kinase (JNK), and so on. Melatonin 51-60 heme oxygenase 1 Homo sapiens 99-115 25033992-6 2014 Thereafter, we introduce some information covering melatonin-related signaling pathways, including heme oxygenase-1 (HO-1), toll-like receptor (TLR), c-Jun N-terminal kinase (JNK), and so on. Melatonin 51-60 heme oxygenase 1 Homo sapiens 117-121 25090972-7 2014 NADA enhances angiogenesis in endothelial vascular cells and promotes the expression of genes such as erythropoietin (EPO), vascular endothelial growth factor A (VEGFA), heme oxygenase 1 (HMOX-1), hexokinase 2 (HK2) and Bcl-2/E1B-nineteen kiloDalton interacting protein (BNIP3) in primary astrocytes. arachidonyl dopamine 0-4 heme oxygenase 1 Homo sapiens 170-186 25090972-7 2014 NADA enhances angiogenesis in endothelial vascular cells and promotes the expression of genes such as erythropoietin (EPO), vascular endothelial growth factor A (VEGFA), heme oxygenase 1 (HMOX-1), hexokinase 2 (HK2) and Bcl-2/E1B-nineteen kiloDalton interacting protein (BNIP3) in primary astrocytes. arachidonyl dopamine 0-4 heme oxygenase 1 Homo sapiens 188-194 23649983-8 2014 Also, treatment of the vapor samples with glutathione resulted in reduction in the Nrf2 activation and HO-1 induction, suggesting that electrophiles in vapor samples contribute to this Nrf2-dependent antioxidant or adaptive response. Glutathione 42-53 heme oxygenase 1 Homo sapiens 103-107 24651515-0 2014 Delayed remote ischemic preconditioning produces an additive cardioprotection to sevoflurane postconditioning through an enhanced heme oxygenase 1 level partly via nuclear factor erythroid 2-related factor 2 nuclear translocation. Sevoflurane 81-92 heme oxygenase 1 Homo sapiens 130-146 25175370-0 2014 Induction of heme oxygenase-1 expression protects articular chondrocytes against cilostazol-induced cellular senescence. Cilostazol 81-91 heme oxygenase 1 Homo sapiens 13-29 25175370-5 2014 Cilostazol also significantly induced HO-1 expression, and the induction of HO-1 expression was affected by a significant increase in reactive oxygen species (ROS) production caused by cilostazol treatment. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 38-42 25175370-5 2014 Cilostazol also significantly induced HO-1 expression, and the induction of HO-1 expression was affected by a significant increase in reactive oxygen species (ROS) production caused by cilostazol treatment. Reactive Oxygen Species 134-157 heme oxygenase 1 Homo sapiens 76-80 25175370-5 2014 Cilostazol also significantly induced HO-1 expression, and the induction of HO-1 expression was affected by a significant increase in reactive oxygen species (ROS) production caused by cilostazol treatment. Reactive Oxygen Species 159-162 heme oxygenase 1 Homo sapiens 76-80 25175370-5 2014 Cilostazol also significantly induced HO-1 expression, and the induction of HO-1 expression was affected by a significant increase in reactive oxygen species (ROS) production caused by cilostazol treatment. Cilostazol 185-195 heme oxygenase 1 Homo sapiens 76-80 25175370-6 2014 Of note, pre-treatment with 3-morpholinosydnonimine hydrochloride (SIN-1), an inducer of HO-1 expression, markedly attenuated cilostazol-induced chondrocyte senescence, and thus, we examined whether HO-1 directly modulates chondrocyte senescence induced by cilostazol. linsidomine 28-65 heme oxygenase 1 Homo sapiens 89-93 25175370-6 2014 Of note, pre-treatment with 3-morpholinosydnonimine hydrochloride (SIN-1), an inducer of HO-1 expression, markedly attenuated cilostazol-induced chondrocyte senescence, and thus, we examined whether HO-1 directly modulates chondrocyte senescence induced by cilostazol. Cilostazol 126-136 heme oxygenase 1 Homo sapiens 89-93 25175370-6 2014 Of note, pre-treatment with 3-morpholinosydnonimine hydrochloride (SIN-1), an inducer of HO-1 expression, markedly attenuated cilostazol-induced chondrocyte senescence, and thus, we examined whether HO-1 directly modulates chondrocyte senescence induced by cilostazol. Cilostazol 126-136 heme oxygenase 1 Homo sapiens 199-203 25175370-7 2014 The upregulation of HO-1 was found to suppress cilostazol-induced cellular senescence. Cilostazol 47-57 heme oxygenase 1 Homo sapiens 20-24 25175370-8 2014 In addition, the inhibition of HO-1 activity with the iron chelator, desferrioxamine (DFO), or HO-1 siRNA increased cilostazol-induced chondrocyte senescence. Iron 54-58 heme oxygenase 1 Homo sapiens 31-35 25175370-8 2014 In addition, the inhibition of HO-1 activity with the iron chelator, desferrioxamine (DFO), or HO-1 siRNA increased cilostazol-induced chondrocyte senescence. Deferoxamine 69-84 heme oxygenase 1 Homo sapiens 31-35 25175370-8 2014 In addition, the inhibition of HO-1 activity with the iron chelator, desferrioxamine (DFO), or HO-1 siRNA increased cilostazol-induced chondrocyte senescence. Deferoxamine 86-89 heme oxygenase 1 Homo sapiens 31-35 25175370-8 2014 In addition, the inhibition of HO-1 activity with the iron chelator, desferrioxamine (DFO), or HO-1 siRNA increased cilostazol-induced chondrocyte senescence. Cilostazol 116-126 heme oxygenase 1 Homo sapiens 31-35 25175370-8 2014 In addition, the inhibition of HO-1 activity with the iron chelator, desferrioxamine (DFO), or HO-1 siRNA increased cilostazol-induced chondrocyte senescence. Cilostazol 116-126 heme oxygenase 1 Homo sapiens 95-99 25172557-7 2014 In addition to its action on the reporter constructs, mRNA and protein levels of heme oxygenase 1, an endogenous target gene of Nrf2, was markedly upregulated by E2 both alone and in combination with tBHQ. 2-tert-butylhydroquinone 200-204 heme oxygenase 1 Homo sapiens 81-97 24762402-1 2014 The length polymorphism of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter is associated with cardiovascular events and mortality in high-risk populations. guanosine thymidine dinucleotide 27-59 heme oxygenase 1 Homo sapiens 75-91 24762402-4 2014 We analyzed the allelic frequencies of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter in 386 patients with coronary artery disease recruited from January 1999 to July 2001 and followed until August 31, 2012. guanosine thymidine dinucleotide 39-71 heme oxygenase 1 Homo sapiens 87-103 24762402-10 2014 In conclusion, a greater number of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter is associated with higher risk for CKD, cardiovascular events, and mortality among patients with coronary artery disease. guanosine thymidine dinucleotide 35-67 heme oxygenase 1 Homo sapiens 83-99 24651515-9 2014 Hence, we conclude that DRIPC may produce an additive cardioprotection to SPoC through an enhanced HO-1 expression partly via Nrf2 translocation. dripc 24-29 heme oxygenase 1 Homo sapiens 99-103 24968709-0 2014 Epigallocatechin-3-gallate (EGCG) protects skin cells from ionizing radiation via heme oxygenase-1 (HO-1) overexpression. epigallocatechin gallate 0-26 heme oxygenase 1 Homo sapiens 82-98 24968709-0 2014 Epigallocatechin-3-gallate (EGCG) protects skin cells from ionizing radiation via heme oxygenase-1 (HO-1) overexpression. epigallocatechin gallate 0-26 heme oxygenase 1 Homo sapiens 100-104 24968709-0 2014 Epigallocatechin-3-gallate (EGCG) protects skin cells from ionizing radiation via heme oxygenase-1 (HO-1) overexpression. epigallocatechin gallate 28-32 heme oxygenase 1 Homo sapiens 82-98 24968709-0 2014 Epigallocatechin-3-gallate (EGCG) protects skin cells from ionizing radiation via heme oxygenase-1 (HO-1) overexpression. epigallocatechin gallate 28-32 heme oxygenase 1 Homo sapiens 100-104 24968709-9 2014 Furthermore, EGCG induced the expression of the cytoprotective molecule heme oxygenase-1 (HO-1) in a dose-dependent manner via transcriptional activation. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 72-88 24968709-9 2014 Furthermore, EGCG induced the expression of the cytoprotective molecule heme oxygenase-1 (HO-1) in a dose-dependent manner via transcriptional activation. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 90-94 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. tin protoporphyrin IX 52-70 heme oxygenase 1 Homo sapiens 37-41 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. tin protoporphyrin IX 52-70 heme oxygenase 1 Homo sapiens 37-41 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. tin protoporphyrin IX 72-78 heme oxygenase 1 Homo sapiens 37-41 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. tin protoporphyrin IX 72-78 heme oxygenase 1 Homo sapiens 37-41 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. epigallocatechin gallate 112-116 heme oxygenase 1 Homo sapiens 0-4 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. epigallocatechin gallate 112-116 heme oxygenase 1 Homo sapiens 37-41 24968709-10 2014 HO-1 knockdown or treatment with the HO-1 inhibitor tin protoporphyrin (SnPPIX) reversed the protective role of EGCG, indicating an important role for HO-1. epigallocatechin gallate 112-116 heme oxygenase 1 Homo sapiens 37-41 25036905-2 2014 In this study, we hypothesized that the expression of heme oxygenase-1 determined by the number of guanidinium thiocyanate (GT) repeats can influence the occurrence of acute pancreatitis. Guanidine 99-110 heme oxygenase 1 Homo sapiens 54-70 25036905-2 2014 In this study, we hypothesized that the expression of heme oxygenase-1 determined by the number of guanidinium thiocyanate (GT) repeats can influence the occurrence of acute pancreatitis. thiocyanate 111-122 heme oxygenase 1 Homo sapiens 54-70 25156196-11 2014 It was also demonstrated that HO-1 is modulated by NO, as shown by experiments performed in the presence of an NO donor (sodium nitroprusside), an NO scavenger (2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide), or an NO synthesis inhibitor (N-nitro-l-arginine methyl ester, NAME). Nitroprusside 121-141 heme oxygenase 1 Homo sapiens 30-34 25213834-4 2014 Knockdown of Nrf2 by siRNA (siNrf2) targeting caused additional refractoriness of HO-1 protein induction to a second UVA or heme treatment and this treatment also further enhanced cell damage by a second dose of UVA radiation. Heme 124-128 heme oxygenase 1 Homo sapiens 82-86 25156196-11 2014 It was also demonstrated that HO-1 is modulated by NO, as shown by experiments performed in the presence of an NO donor (sodium nitroprusside), an NO scavenger (2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide), or an NO synthesis inhibitor (N-nitro-l-arginine methyl ester, NAME). 1,3-dihydroxy-4,4,5,5-tetramethyl-2-(4-carboxyphenyl)tetrahydroimidazole 161-227 heme oxygenase 1 Homo sapiens 30-34 25156196-11 2014 It was also demonstrated that HO-1 is modulated by NO, as shown by experiments performed in the presence of an NO donor (sodium nitroprusside), an NO scavenger (2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide), or an NO synthesis inhibitor (N-nitro-l-arginine methyl ester, NAME). n-nitro-l-arginine methyl ester 259-290 heme oxygenase 1 Homo sapiens 30-34 25190803-8 2014 The mammalian target of rapamycin complex 2 (mTORC2) inhibitor, AZD2014, ablated XBP1u or HDAC3 or disturbed flow-mediated Akt1 phosphorylation, Nrf2 nuclear translocation, and HO-1 expression. vistusertib 64-71 heme oxygenase 1 Homo sapiens 177-181 25105464-6 2014 Treatment with CDDO-Me increased the gene expression of NQO1 (P = 0.04) and decreased the expression of HO-1 (P = 0.03) in PBMCs from patients with septic shock. bardoxolone methyl 15-22 heme oxygenase 1 Homo sapiens 104-108 25169430-4 2014 Co-treatment with 100muM sesamin for 12h up-regulated the HO-1 protein level increased by LPS; however, HO-1 mRNA was unaffected. sesamin 25-32 heme oxygenase 1 Homo sapiens 58-62 25223958-0 2014 Upregulation of both heme oxygenase-1 and ATPase inhibitory factor 1 renders tumoricidal activity by synthetic flavonoids via depleting cellular ATP. Flavonoids 111-121 heme oxygenase 1 Homo sapiens 21-37 25169430-5 2014 Sesamin delayed the reversal, by the protein synthesis inhibitor cycloheximide (1muM), of the LPS-induced increase of HO-1 protein level. sesamin 0-7 heme oxygenase 1 Homo sapiens 118-122 25223958-8 2014 To gain a physiological significance, we treated the synthetic flavonoids in colon cancer cells, HT29 and HCT116 cells and confirmed that overexpression of both HO-1 and ATPIF1 was critical for tumor cell death with an impaired mitochondrial energetics. Flavonoids 63-73 heme oxygenase 1 Homo sapiens 161-165 25169430-10 2014 Co-treatment with sesamin decreased ubiquitinated HO-1 protein accumulation by MG-132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 79-85 heme oxygenase 1 Homo sapiens 50-54 25169430-12 2014 These findings suggest that sesamin disturbs the degradation of HO-1 protein through inhibiting its ubiquitination, resulting in HO-1 protein up-regulation. sesamin 28-35 heme oxygenase 1 Homo sapiens 64-68 25169430-5 2014 Sesamin delayed the reversal, by the protein synthesis inhibitor cycloheximide (1muM), of the LPS-induced increase of HO-1 protein level. Cycloheximide 65-78 heme oxygenase 1 Homo sapiens 118-122 25169430-12 2014 These findings suggest that sesamin disturbs the degradation of HO-1 protein through inhibiting its ubiquitination, resulting in HO-1 protein up-regulation. sesamin 28-35 heme oxygenase 1 Homo sapiens 129-133 25169430-8 2014 To clarify the mechanisms that underlie the up-regulation of HO-1 protein level by sesamin, the human embryonic kidney (HEK) 293T cell line transfected with Flag-tagged HO-1 was used. sesamin 83-90 heme oxygenase 1 Homo sapiens 61-65 25169430-9 2014 A proteasome inhibitor, MG-132 (10muM), stabilized HO-1 protein in HEK 293T cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 24-30 heme oxygenase 1 Homo sapiens 51-55 25169430-10 2014 Co-treatment with sesamin decreased ubiquitinated HO-1 protein accumulation by MG-132. sesamin 18-25 heme oxygenase 1 Homo sapiens 50-54 24499676-4 2014 In addition, the transduced PEP-1-HO-1 inhibited generation of reactive oxygen species (ROS) and cell death caused by 1-methyl-4-phenylpyridinium ion (MPP+). Reactive Oxygen Species 63-86 heme oxygenase 1 Homo sapiens 34-38 25325059-8 2014 Induction of heme oxygenase-1, which can be attained with phase 2 inducer phytochemicals such as lipoic acid and green tea catechins, promotes reverse cholesterol transport in macrophages and inhibits atherogenesis in rodents, likely due to, in large part, NF-kappaB antagonism. Thioctic Acid 97-108 heme oxygenase 1 Homo sapiens 13-29 25325059-8 2014 Induction of heme oxygenase-1, which can be attained with phase 2 inducer phytochemicals such as lipoic acid and green tea catechins, promotes reverse cholesterol transport in macrophages and inhibits atherogenesis in rodents, likely due to, in large part, NF-kappaB antagonism. Catechin 123-132 heme oxygenase 1 Homo sapiens 13-29 25325059-8 2014 Induction of heme oxygenase-1, which can be attained with phase 2 inducer phytochemicals such as lipoic acid and green tea catechins, promotes reverse cholesterol transport in macrophages and inhibits atherogenesis in rodents, likely due to, in large part, NF-kappaB antagonism. Cholesterol 151-162 heme oxygenase 1 Homo sapiens 13-29 25086213-2 2014 Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Heme 112-116 heme oxygenase 1 Homo sapiens 0-16 25086213-2 2014 Heme oxygenase-1 (HO-1) is a ubiquitously expressed inducible isoform of the first and rate-limiting enzyme for heme degradation. Heme 112-116 heme oxygenase 1 Homo sapiens 18-22 25341814-0 2014 Atorvastatin Attenuates TNF-alpha Production via Heme Oxygenase-1 Pathway in LPS-stimulated RAW264.7 Macrophages. Atorvastatin 0-12 heme oxygenase 1 Homo sapiens 49-65 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 76-88 heme oxygenase 1 Homo sapiens 4-8 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 76-88 heme oxygenase 1 Homo sapiens 22-26 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 111-123 heme oxygenase 1 Homo sapiens 4-8 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 111-123 heme oxygenase 1 Homo sapiens 22-26 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. SB 203580 152-160 heme oxygenase 1 Homo sapiens 4-8 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. SB 203580 152-160 heme oxygenase 1 Homo sapiens 22-26 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 165-172 heme oxygenase 1 Homo sapiens 4-8 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 165-172 heme oxygenase 1 Homo sapiens 22-26 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 111-123 heme oxygenase 1 Homo sapiens 4-8 25341814-7 2014 The HO-1 activity and HO-1 expression level were significantly higher after atorvastatin treatment than before atorvastatin treatment and attenuated by SB203580 and PD98059 but not by SP600125, suggesting that the ERK and p38 mitogen-activated protein kinase (MAPK) pathways participate in regulating the above-mentioned effects of atorvastatin. Atorvastatin 111-123 heme oxygenase 1 Homo sapiens 22-26 25341814-8 2014 Moreover, the HO-1 activity suppressed by SnPP or the HO-1 expression inhibited by siRNA significantly attenuated the effect of atorvastatin on TNF-alpha expression and production in LPS-stimulated macrophages. Atorvastatin 128-140 heme oxygenase 1 Homo sapiens 14-18 25341814-8 2014 Moreover, the HO-1 activity suppressed by SnPP or the HO-1 expression inhibited by siRNA significantly attenuated the effect of atorvastatin on TNF-alpha expression and production in LPS-stimulated macrophages. Atorvastatin 128-140 heme oxygenase 1 Homo sapiens 54-58 25341814-9 2014 CONCLUSION: Atorvastatin can attenuate LPS-induced TNF-alpha expression and production by activating HO-1 via the ERK and p38 MAPK pathways, suggesting that atorvastatin can be used in treatment of inflammatory diseases such as sepsis, especially in those with atherosclerotic diseases. Atorvastatin 12-24 heme oxygenase 1 Homo sapiens 101-105 25341814-9 2014 CONCLUSION: Atorvastatin can attenuate LPS-induced TNF-alpha expression and production by activating HO-1 via the ERK and p38 MAPK pathways, suggesting that atorvastatin can be used in treatment of inflammatory diseases such as sepsis, especially in those with atherosclerotic diseases. Atorvastatin 157-169 heme oxygenase 1 Homo sapiens 101-105 24934751-0 2014 AP-1 activation attenuates the arsenite-induced apoptotic response in human bronchial epithelial cells by up-regulating HO-1 expression. arsenite 31-39 heme oxygenase 1 Homo sapiens 120-124 24934751-4 2014 Here, we show that a significant induction of HO-1 expression is present in human bronchial epithelial cells (Beas-2B) treated with lethal doses of arsenite treatment. arsenite 148-156 heme oxygenase 1 Homo sapiens 46-50 24934751-6 2014 As expected, HO-1 RNAi knockdown, or ERK/AP1 inhibition, renders the Beas-2B cells more sensitive to arsenite damage. arsenite 101-109 heme oxygenase 1 Homo sapiens 13-17 24934751-7 2014 Our data thus suggest that transcriptional upregulation of HO-1 expression via a putative ERK/AP-1 pathway constitutes an inherent mechanism by which arsenite-induced apoptosis is attenuated. arsenite 150-158 heme oxygenase 1 Homo sapiens 59-63 24499676-4 2014 In addition, the transduced PEP-1-HO-1 inhibited generation of reactive oxygen species (ROS) and cell death caused by 1-methyl-4-phenylpyridinium ion (MPP+). Reactive Oxygen Species 88-91 heme oxygenase 1 Homo sapiens 34-38 24499676-4 2014 In addition, the transduced PEP-1-HO-1 inhibited generation of reactive oxygen species (ROS) and cell death caused by 1-methyl-4-phenylpyridinium ion (MPP+). 1-Methyl-4-phenylpyridinium 118-149 heme oxygenase 1 Homo sapiens 34-38 24499676-4 2014 In addition, the transduced PEP-1-HO-1 inhibited generation of reactive oxygen species (ROS) and cell death caused by 1-methyl-4-phenylpyridinium ion (MPP+). mangion-purified polysaccharide (Candida albicans) 151-155 heme oxygenase 1 Homo sapiens 34-38 24732176-3 2014 Such investigation could be important, as iron and carbon monoxide are two of the products of heme catabolism via heme oxygenase-1, an enzyme upregulated in a variety of disease states associated with thrombophilia. Iron 42-46 heme oxygenase 1 Homo sapiens 114-130 24732176-3 2014 Such investigation could be important, as iron and carbon monoxide are two of the products of heme catabolism via heme oxygenase-1, an enzyme upregulated in a variety of disease states associated with thrombophilia. Carbon Monoxide 51-66 heme oxygenase 1 Homo sapiens 114-130 24732176-3 2014 Such investigation could be important, as iron and carbon monoxide are two of the products of heme catabolism via heme oxygenase-1, an enzyme upregulated in a variety of disease states associated with thrombophilia. Heme 94-98 heme oxygenase 1 Homo sapiens 114-130 25150395-0 2014 Up-regulation of heme oxygenase-1 expression modulates reactive oxygen species level during the cryopreservation of human seminiferous tubules. Reactive Oxygen Species 55-78 heme oxygenase 1 Homo sapiens 17-33 25150395-13 2014 The ROS level was negatively correlated with HO-1 expression. Reactive Oxygen Species 4-7 heme oxygenase 1 Homo sapiens 45-49 25023856-2 2014 Described here are studies of tyrosyl radical formation reactions that occur after oxidizing Fe(III)(Por) to Fe(IV)=O(Por( +)) in human heme oxygenase isoform-1 (hHO-1) and the structurally homologous protein from Corynebacterium diphtheriae (cdHO). Tyrosyl radical 30-45 heme oxygenase 1 Homo sapiens 162-167 24993616-7 2014 Furthermore, sulforaphane was observed to activate endoplasmic reticulum (ER) stress and the nuclear factor-E2-related factor-2 (Nrf2) signaling pathway, as demonstrated by the upregulation of ER stress-related proteins, including glucose-regulated protein 78 and C/EBP-homologous protein, and the accumulation of phosphorylated Nrf2 proteins in the nucleus and induction of heme oxygenase-1 expression, respectively. sulforaphane 13-25 heme oxygenase 1 Homo sapiens 375-391 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). sulforaphane 0-12 heme oxygenase 1 Homo sapiens 126-142 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). sulforaphane 0-12 heme oxygenase 1 Homo sapiens 144-148 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). sulforaphane 14-17 heme oxygenase 1 Homo sapiens 126-142 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). sulforaphane 14-17 heme oxygenase 1 Homo sapiens 144-148 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). isothiocyanic acid 25-39 heme oxygenase 1 Homo sapiens 126-142 26461289-4 2014 Sulforaphane (SFN) is an isothiocyanate present in cruciferous vegetables that can induce antioxidant defence enzymes such as heme oxygenase-1 (HO-1) and peroxiredoxin-1 (Prx-1). isothiocyanic acid 25-39 heme oxygenase 1 Homo sapiens 144-148 25091622-0 2014 Tussilagone inhibits dendritic cell functions via induction of heme oxygenase-1. tussilagone 0-11 heme oxygenase 1 Homo sapiens 63-79 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. tussilagone 40-43 heme oxygenase 1 Homo sapiens 54-70 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. tussilagone 40-43 heme oxygenase 1 Homo sapiens 72-76 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. Heme 54-58 heme oxygenase 1 Homo sapiens 72-76 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. Carbon Monoxide 138-153 heme oxygenase 1 Homo sapiens 54-70 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. Carbon Monoxide 138-153 heme oxygenase 1 Homo sapiens 72-76 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. Bilirubin 158-167 heme oxygenase 1 Homo sapiens 54-70 25091622-5 2014 As a critical mechanism, we showed that TUS activated heme oxygenase-1 (HO-1), which degrades heme to immunosuppressive products, such as carbon monoxide and bilirubin. Bilirubin 158-167 heme oxygenase 1 Homo sapiens 72-76 25091622-6 2014 HO-1 inhibitor reversed the inhibitory activity of TUS in DCs. tussilagone 51-54 heme oxygenase 1 Homo sapiens 0-4 25091622-7 2014 In conclusion, this study suggests that TUS inhibits DC function through the induction of HO-1. tussilagone 40-43 heme oxygenase 1 Homo sapiens 90-94 25091623-5 2014 However, 3-DSC-mediated HO-1 induction was completely blocked by treatment with cycloheximide, a translational inhibitor, or rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR). Cycloheximide 80-93 heme oxygenase 1 Homo sapiens 24-28 25091623-5 2014 However, 3-DSC-mediated HO-1 induction was completely blocked by treatment with cycloheximide, a translational inhibitor, or rapamycin, an inhibitor of the mammalian target of rapamycin (mTOR). Sirolimus 125-134 heme oxygenase 1 Homo sapiens 24-28 25202977-6 2014 HO-1 siRNA knockdown and HO enzymatic inhibition in HIV-infected macrophages increased supernatant glutamate and neurotoxicity. Glutamic Acid 99-108 heme oxygenase 1 Homo sapiens 0-4 25202977-7 2014 In contrast, increasing HO-1 expression through siRNA derepression or with nonselective pharmacologic inducers, including the CNS-penetrating drug dimethyl fumarate (DMF), decreased supernatant glutamate and neurotoxicity. Dimethyl Fumarate 147-164 heme oxygenase 1 Homo sapiens 24-28 25202977-7 2014 In contrast, increasing HO-1 expression through siRNA derepression or with nonselective pharmacologic inducers, including the CNS-penetrating drug dimethyl fumarate (DMF), decreased supernatant glutamate and neurotoxicity. Dimethyl Fumarate 166-169 heme oxygenase 1 Homo sapiens 24-28 25202977-7 2014 In contrast, increasing HO-1 expression through siRNA derepression or with nonselective pharmacologic inducers, including the CNS-penetrating drug dimethyl fumarate (DMF), decreased supernatant glutamate and neurotoxicity. Glutamic Acid 194-203 heme oxygenase 1 Homo sapiens 24-28 25023856-2 2014 Described here are studies of tyrosyl radical formation reactions that occur after oxidizing Fe(III)(Por) to Fe(IV)=O(Por( +)) in human heme oxygenase isoform-1 (hHO-1) and the structurally homologous protein from Corynebacterium diphtheriae (cdHO). ferric sulfate 93-100 heme oxygenase 1 Homo sapiens 162-167 25023856-2 2014 Described here are studies of tyrosyl radical formation reactions that occur after oxidizing Fe(III)(Por) to Fe(IV)=O(Por( +)) in human heme oxygenase isoform-1 (hHO-1) and the structurally homologous protein from Corynebacterium diphtheriae (cdHO). fe(iv) 109-115 heme oxygenase 1 Homo sapiens 162-167 25023856-3 2014 Site-directed mutagenesis on hHO-1 probes the reduction of Fe(IV)=O(Por( +)) by tyrosine residues within 11 A of the prosthetic group. Tyrosine 80-88 heme oxygenase 1 Homo sapiens 29-34 24734887-6 2014 We found that NAC significantly increased Nrf2 and downstream HO-1 expression. Acetylcysteine 14-17 heme oxygenase 1 Homo sapiens 62-66 25037998-5 2014 In vitro, caffeic acid increases the nuclear/cytosolic Nrf2 ratio and the mRNA expression of the downstream antioxidant enzymes, Upsilon-glutamyl cysteine synthetase, glutathione peroxidase, glutathione reductase, and heme oxygenase-1, by activating the JNK/p38 pathway in Int-407 cells. caffeic acid 10-22 heme oxygenase 1 Homo sapiens 218-234 25093998-0 2014 Effects of aspirin & simvastatin and aspirin, simvastatin, & lipoic acid on heme oxygenase-1 in healthy human subjects. Aspirin 41-48 heme oxygenase 1 Homo sapiens 84-100 25093998-0 2014 Effects of aspirin & simvastatin and aspirin, simvastatin, & lipoic acid on heme oxygenase-1 in healthy human subjects. Adenosine Monophosphate 64-67 heme oxygenase 1 Homo sapiens 84-100 25093998-0 2014 Effects of aspirin & simvastatin and aspirin, simvastatin, & lipoic acid on heme oxygenase-1 in healthy human subjects. Thioctic Acid 69-80 heme oxygenase 1 Homo sapiens 84-100 25093998-3 2014 However, the only drugs (i.e., hemin and heme arginate) which pharmacologically upregulate HO-1 in humans are expensive and can only be administered intravenously. Hemin 31-36 heme oxygenase 1 Homo sapiens 91-95 25093998-3 2014 However, the only drugs (i.e., hemin and heme arginate) which pharmacologically upregulate HO-1 in humans are expensive and can only be administered intravenously. heme arginate 41-54 heme oxygenase 1 Homo sapiens 91-95 25093998-4 2014 Our aims were to compare the effects of placebo, aspirin, and simvastatin alone, and with alpha-lipoic acid, on HO-1 protein concentration and activity in humans. Simvastatin 62-73 heme oxygenase 1 Homo sapiens 112-116 25093998-4 2014 Our aims were to compare the effects of placebo, aspirin, and simvastatin alone, and with alpha-lipoic acid, on HO-1 protein concentration and activity in humans. Thioctic Acid 90-107 heme oxygenase 1 Homo sapiens 112-116 25309329-5 2014 The isoform HMOX1 is highly inducible in response to reactive oxygen species, which induce an increase in BBB permeability and impair its pathophysiology. Reactive Oxygen Species 53-76 heme oxygenase 1 Homo sapiens 12-17 25355252-5 2014 Compared with the control group, the serum HO-1 was significantly increased at 6, 12, 24, 48 h after the heart valve replacement surgery in both the RIPerC and RIPostC groups (P<0.05); the SCr, BUN, urinary NGAL and serum iron values were decreased at 6, 12, 24, 48 h after the heart valve replacement surgery in both the RIPerC and RIPostC groups (P>0.05). Iron 225-229 heme oxygenase 1 Homo sapiens 43-47 25107906-5 2014 This regulation of Nrf2 post-induction by nuclear HO-1 is important for the preferential transcription of phase II detoxification enzymes such as NQO1 as well as glucose-6-phosphate dehydrogenase (G6PDH), a regulator of the pentose phosphate pathway. Pentosephosphates 224-241 heme oxygenase 1 Homo sapiens 50-54 25272046-0 2014 Long-term aerobic exercise protects against cisplatin-induced nephrotoxicity by modulating the expression of IL-6 and HO-1. Cisplatin 44-53 heme oxygenase 1 Homo sapiens 118-122 25148906-4 2014 Further analysis using gene microarray showed that 6S upregulated the expression of Nrf2 target genes (AKR1B10, FTL, GGTLA4, and HMOX1) in HCT-116 cells. S 6 51-53 heme oxygenase 1 Homo sapiens 129-134 24954650-7 2014 HO-1 metabolizes heme to biliverdin, iron and carbon monoxide (CO). Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 25182732-5 2014 The collective application of polyunsaturated fatty acids (PUFAs) and irradiation significantly changed the expression of EGR1, TNF-alpha, NOTCH1, c-MYC, TP53, HMOX1, AKR1C1, NQO1, while up-regulation of GADD45A, EGR1, GRP78, DDIT3, c-MYC, FOSL1 were recorded both in response to PUFA treatment or irradiation alone. Fatty Acids, Unsaturated 30-57 heme oxygenase 1 Homo sapiens 160-165 24954650-7 2014 HO-1 metabolizes heme to biliverdin, iron and carbon monoxide (CO). Biliverdine 25-35 heme oxygenase 1 Homo sapiens 0-4 24954650-7 2014 HO-1 metabolizes heme to biliverdin, iron and carbon monoxide (CO). Iron 37-41 heme oxygenase 1 Homo sapiens 0-4 24954650-7 2014 HO-1 metabolizes heme to biliverdin, iron and carbon monoxide (CO). Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 0-4 24954650-7 2014 HO-1 metabolizes heme to biliverdin, iron and carbon monoxide (CO). Carbon Monoxide 63-65 heme oxygenase 1 Homo sapiens 0-4 24998495-0 2014 Andrographolide inhibits TNFalpha-induced ICAM-1 expression via suppression of NADPH oxidase activation and induction of HO-1 and GCLM expression through the PI3K/Akt/Nrf2 and PI3K/Akt/AP-1 pathways in human endothelial cells. andrographolide 0-15 heme oxygenase 1 Homo sapiens 121-125 24998495-7 2014 Andrographolide induced the gene expression of heme oxygenase 1 (HO-1) and glutamate cysteine ligase modifier subunit (GCLM) in a time-dependent manner. andrographolide 0-15 heme oxygenase 1 Homo sapiens 47-63 24998495-7 2014 Andrographolide induced the gene expression of heme oxygenase 1 (HO-1) and glutamate cysteine ligase modifier subunit (GCLM) in a time-dependent manner. andrographolide 0-15 heme oxygenase 1 Homo sapiens 65-69 24998495-11 2014 The mechanism underlying the up-regulation of HO-1 and GCLM by andrographolide was dependent on the PI3K/Akt pathway, and both the Nrf2 and AP-1 transcriptional factors were involved. andrographolide 63-78 heme oxygenase 1 Homo sapiens 46-50 24998495-12 2014 Our results suggest that andrographolide attenuates TNFalpha-induced ICAM-1 expression at least partially through suppression of NADPH oxidase activation and induction of HO-1 and GCLM expression, which is PI3K/Akt pathway-dependent. andrographolide 25-40 heme oxygenase 1 Homo sapiens 171-175 24970682-9 2014 Metformin reduced heme oxygenase-1 and superoxide dismutase 2 but upregulated catalase expression in MCF-7 cells. Metformin 0-9 heme oxygenase 1 Homo sapiens 18-34 24568186-0 2014 15-deoxy-Delta12,14-prostaglandin J2 induces p53 expression through Nrf2-mediated upregulation of heme oxygenase-1 in human breast cancer cells. 14-prostaglandin j2 17-36 heme oxygenase 1 Homo sapiens 98-114 25072782-3 2014 CO is a byproduct of heme degradation mediated by heme oxygenase (HO-1). Heme 21-25 heme oxygenase 1 Homo sapiens 66-70 24568186-3 2014 In the present work, we investigated the effects of HO-1 on the expression of p53 induced by 15-deoxy-Delta(12,14)-prostaglandin J2 (15d-PGJ2) in human breast cancer (MCF-7) cells. 9-deoxy-delta-9-prostaglandin D2 115-131 heme oxygenase 1 Homo sapiens 52-56 24568186-4 2014 Treatment of MCF-7 cells with 15d-PGJ2 led to time-dependent increases in the expression of p53 as well as HO-1. 15-deoxy-delta(12,14)-prostaglandin J2 30-38 heme oxygenase 1 Homo sapiens 107-111 24568186-5 2014 Upregulation of p53 expression by 15d-PGJ2 was abrogated by si-RNA knock-down of HO-1. 15-deoxy-delta(12,14)-prostaglandin J2 34-42 heme oxygenase 1 Homo sapiens 81-85 24568186-8 2014 We speculated that iron, a by-product of HO-1-catalyzed reactions, could mediate 15d-PGJ2-induced p53 expression. Iron 19-23 heme oxygenase 1 Homo sapiens 41-45 24568186-8 2014 We speculated that iron, a by-product of HO-1-catalyzed reactions, could mediate 15d-PGJ2-induced p53 expression. 15-deoxy-delta(12,14)-prostaglandin J2 81-89 heme oxygenase 1 Homo sapiens 41-45 24568186-10 2014 Iron released from heme by HO-1 activity is mostly in the Fe(2+) form. Iron 0-4 heme oxygenase 1 Homo sapiens 27-31 24568186-10 2014 Iron released from heme by HO-1 activity is mostly in the Fe(2+) form. Heme 19-23 heme oxygenase 1 Homo sapiens 27-31 24568186-10 2014 Iron released from heme by HO-1 activity is mostly in the Fe(2+) form. ammonium ferrous sulfate 58-64 heme oxygenase 1 Homo sapiens 27-31 24932697-0 2014 Gastrodin protects against MPP(+)-induced oxidative stress by up regulates heme oxygenase-1 expression through p38 MAPK/Nrf2 pathway in human dopaminergic cells. gastrodin 0-9 heme oxygenase 1 Homo sapiens 75-91 24932697-8 2014 And the increase in HO-1 expression was correlated with the protective effect of gastrodin against MPP(+)-induced injury. gastrodin 81-90 heme oxygenase 1 Homo sapiens 20-24 24932697-8 2014 And the increase in HO-1 expression was correlated with the protective effect of gastrodin against MPP(+)-induced injury. mangion-purified polysaccharide (Candida albicans) 99-105 heme oxygenase 1 Homo sapiens 20-24 24932697-9 2014 Because the inhibitor of HO-1 activity, ZnPP reversed the protective effect of gastrodin against MPP(+)-induced cell death. zinc protoporphyrin 40-44 heme oxygenase 1 Homo sapiens 25-29 24932697-9 2014 Because the inhibitor of HO-1 activity, ZnPP reversed the protective effect of gastrodin against MPP(+)-induced cell death. gastrodin 79-88 heme oxygenase 1 Homo sapiens 25-29 24932697-10 2014 We also demonstrated that the specific p38 MAPK inhibitor, SB203580, concentration-dependently blocked on gastrodin-induced HO-1 expression, and meanwhile SB203580 reversed the protective effect of gastrodin against MPP(+)-induced cell death. SB 203580 59-67 heme oxygenase 1 Homo sapiens 124-128 24932697-11 2014 Taken together, these findings suggest that gastrodin can induce HO-1 expression through activation of p38 MAPK/Nrf2 signaling pathway, thereby protecting the SH-SY5Y cells from MPP(+)-induced oxidative cell death. gastrodin 44-53 heme oxygenase 1 Homo sapiens 65-69 24963040-3 2014 Here, we report that subablative bone marrow transplantation (BMT) has a curative effect for disease in Hmox1(-/-) animals as a result of restoration of heme recycling by repopulation of the tissues with wild-type macrophages. Heme 153-157 heme oxygenase 1 Homo sapiens 104-109 24963040-6 2014 These cells, identified as Kupffer cells with high levels of Hmox1 expression, persisted months after transient engraftment of the donor bone marrow and were responsible for the full restoration of heme-recycling ability in Hmox1(-/-) mice and reversing Hmox1-deficient phenotype. Heme 198-202 heme oxygenase 1 Homo sapiens 224-229 24718901-8 2014 WA demonstrated induction of N-acetyl-L-cysteine-repressible oxidative stress as measured directly and through a subsequent heat shock response with HSP32 and HSP70 upregulation and decreased HSF1. Acetylcysteine 29-48 heme oxygenase 1 Homo sapiens 149-154 24662764-3 2014 Our previous study showed that the nuclear factor E2-related factor 2-antioxidant response element (Nrf2-ARE) pathway and its downstream antioxidant enzyme heme oxygenase-1 (HO-1) are crucial for melanocytes to cope with H2O2-induced oxidative damage. Hydrogen Peroxide 221-225 heme oxygenase 1 Homo sapiens 156-172 24662764-3 2014 Our previous study showed that the nuclear factor E2-related factor 2-antioxidant response element (Nrf2-ARE) pathway and its downstream antioxidant enzyme heme oxygenase-1 (HO-1) are crucial for melanocytes to cope with H2O2-induced oxidative damage. Hydrogen Peroxide 221-225 heme oxygenase 1 Homo sapiens 174-178 24662764-5 2014 We found that vitiligo melanocytes exhibited hypersensitivity to H2O2-induced oxidative injury because of reduced Nrf2 nuclear translocation and transcriptional activity, which led to decreased HO-1 expression and aberrant redox balance. Hydrogen Peroxide 65-69 heme oxygenase 1 Homo sapiens 194-198 25371895-0 2014 Sildenafil Inhibits Advanced Glycation End Products-induced sFlt-1 Release Through Upregulation of Heme Oxygenase-1. Sildenafil Citrate 0-10 heme oxygenase 1 Homo sapiens 99-115 25371895-8 2014 CONCLUSION: These results strongly suggest that sildenafil citrate inhibits sFlt-1 release and ROS production in cells treated with AGEs-BSA through upregulation of the HO-1 expression in JEG-3 cells. Sildenafil Citrate 48-66 heme oxygenase 1 Homo sapiens 169-173 25176212-6 2014 The reactive oxygen species were detected in the HO-1-overexpression HEK293T cells and the normal cells after the adding of hydrogen peroxide. Reactive Oxygen Species 4-27 heme oxygenase 1 Homo sapiens 49-53 25176212-6 2014 The reactive oxygen species were detected in the HO-1-overexpression HEK293T cells and the normal cells after the adding of hydrogen peroxide. Hydrogen Peroxide 124-141 heme oxygenase 1 Homo sapiens 49-53 25176212-9 2014 The reactive oxygen species in the HO-1-overexpression HEK293T cells and human umbilical vein endothelial cells decreased obviously after exposure to hydrogen peroxide. Reactive Oxygen Species 4-27 heme oxygenase 1 Homo sapiens 35-39 25176212-9 2014 The reactive oxygen species in the HO-1-overexpression HEK293T cells and human umbilical vein endothelial cells decreased obviously after exposure to hydrogen peroxide. Hydrogen Peroxide 150-167 heme oxygenase 1 Homo sapiens 35-39 25317018-3 2014 To regulate hemeoxygenase-1 (HO-1) expression, the cells were treated CoCl2 or HO-1 siRNA. cobaltous chloride 70-75 heme oxygenase 1 Homo sapiens 12-33 24997245-11 2014 Furthermore, alpha-iso-cubebenol stimulated the expression of multiple antioxidant response genes (NQO-1 and HO-1). alpha-iso-cubebenol 13-32 heme oxygenase 1 Homo sapiens 109-113 25136316-7 2014 In addition, curcumin with its versatile activities modulated the expression of many oxidative stress-regulating genes such as PDGF, VEGF, IGFBP-2, HO1, SOD2, and GPX1. Curcumin 13-21 heme oxygenase 1 Homo sapiens 148-151 25019514-7 2014 Bach1 has an endogenous ligand, heme, that inhibits Bach1 binding to ARE, thus allowing Nrf2-mediated gene expression including that of heme-oxygenase-1 (HMOX1), a well described target of Bach1 repression. Heme 32-36 heme oxygenase 1 Homo sapiens 136-152 25068294-8 2014 Antioxidant proteins including heme oxygenase-1 and peroxiredoxin-2 levels were also increased in retinal AC under high glucose conditions through nuclear localization of transcription factor nuclear factor-erythroid 2-related factor-2. Glucose 120-127 heme oxygenase 1 Homo sapiens 31-47 25019514-7 2014 Bach1 has an endogenous ligand, heme, that inhibits Bach1 binding to ARE, thus allowing Nrf2-mediated gene expression including that of heme-oxygenase-1 (HMOX1), a well described target of Bach1 repression. Heme 32-36 heme oxygenase 1 Homo sapiens 154-159 25007817-0 2014 Nrf2-mediated HO-1 induction coupled with the ERK signaling pathway contributes to indirect antioxidant capacity of caffeic acid phenethyl ester in HepG2 cells. caffeic acid phenethyl ester 116-144 heme oxygenase 1 Homo sapiens 14-18 25170436-7 2014 Results showed a significant increase in the expression of NRF2-responsive genes, GCLC, HMOX1, NQO1, after 1 muM Cd x 1 muM B[a]P co-exposure. Cadmium 113-115 heme oxygenase 1 Homo sapiens 88-93 24981859-3 2014 NO suppresses the pathogenesis of ECM via a mechanism involving (1) the transcription factor nuclear factor erythroid 2-related factor 2 (NRF-2), (2) induction of heme oxygenase-1 (HO-1), and (3) CO production via heme catabolism by HO-1. Heme 163-167 heme oxygenase 1 Homo sapiens 181-185 25007817-4 2014 Treatment with ERK inhibitor U126 completely suppressed CAPE-induced ERK phosphorylation and HO-1 expression, but it only partly inhibited CAPE-induced Nrf2 accumulation and ARE promoter. 3-Bromo-2-fluorotoluene 29-33 heme oxygenase 1 Homo sapiens 93-97 25007817-4 2014 Treatment with ERK inhibitor U126 completely suppressed CAPE-induced ERK phosphorylation and HO-1 expression, but it only partly inhibited CAPE-induced Nrf2 accumulation and ARE promoter. caffeic acid phenethyl ester 56-60 heme oxygenase 1 Homo sapiens 93-97 25007817-6 2014 From the overall results it is proposed that the indirect antioxidant activity of CAPE against oxidative stress in HepG2 cells is partially attributed to induction of HO-1, which is regulated by Kelch-like erythroid-cell-derived protein with CNC homology (ECH)-associated protein 1 (Keap1)-independent Nrf2 activation relying on post-translational phosphorylation of ERK. caffeic acid phenethyl ester 82-86 heme oxygenase 1 Homo sapiens 167-171 24594225-4 2014 RESULTS: 17-Oxo-DHA was a strong inducer of the anti-oxidant response promoting Nrf2 nuclear accumulation, leading to the expression of heme oxygenase 1 and more than doubling glutathione levels. (4Z,7Z,10Z,13Z,15E,19Z)-17-Oxodocosahexaenoic acid 9-19 heme oxygenase 1 Homo sapiens 136-152 24703885-2 2014 We previously reported that caffeic acid phenethyl ester (CAPE), a plant-derived polyphenolic compound, protected human umbilical vein endothelial cells (HUVEC) from menadione-induced oxidative stress and that this cytoprotective effect was correlated with the capacity to induce heme oxygenase-1 (HMOX1) and its protein product, a phase II cytoprotective enzyme. caffeic acid phenethyl ester 28-56 heme oxygenase 1 Homo sapiens 280-296 24703885-2 2014 We previously reported that caffeic acid phenethyl ester (CAPE), a plant-derived polyphenolic compound, protected human umbilical vein endothelial cells (HUVEC) from menadione-induced oxidative stress and that this cytoprotective effect was correlated with the capacity to induce heme oxygenase-1 (HMOX1) and its protein product, a phase II cytoprotective enzyme. caffeic acid phenethyl ester 28-56 heme oxygenase 1 Homo sapiens 298-303 24703885-2 2014 We previously reported that caffeic acid phenethyl ester (CAPE), a plant-derived polyphenolic compound, protected human umbilical vein endothelial cells (HUVEC) from menadione-induced oxidative stress and that this cytoprotective effect was correlated with the capacity to induce heme oxygenase-1 (HMOX1) and its protein product, a phase II cytoprotective enzyme. caffeic acid phenethyl ester 58-62 heme oxygenase 1 Homo sapiens 280-296 24703885-2 2014 We previously reported that caffeic acid phenethyl ester (CAPE), a plant-derived polyphenolic compound, protected human umbilical vein endothelial cells (HUVEC) from menadione-induced oxidative stress and that this cytoprotective effect was correlated with the capacity to induce heme oxygenase-1 (HMOX1) and its protein product, a phase II cytoprotective enzyme. caffeic acid phenethyl ester 58-62 heme oxygenase 1 Homo sapiens 298-303 24703885-6 2014 In addition to potent up-regulation of HMOX1, heat shock proteins (HSP) were also found to be highly induced by CDDO-Im in HUVEC. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 112-119 heme oxygenase 1 Homo sapiens 39-44 24613679-8 2014 RESULTS: rHx-bound heme induced the expression of HO-1 and decreased the level of Bach1 protein. rhx 9-12 heme oxygenase 1 Homo sapiens 50-54 24613679-8 2014 RESULTS: rHx-bound heme induced the expression of HO-1 and decreased the level of Bach1 protein. Heme 19-23 heme oxygenase 1 Homo sapiens 50-54 24613679-9 2014 CPZ inhibited the induction of the HO-1 expression by rHx-bound heme. Chlorpromazine 0-3 heme oxygenase 1 Homo sapiens 35-39 24613679-9 2014 CPZ inhibited the induction of the HO-1 expression by rHx-bound heme. rhx 54-57 heme oxygenase 1 Homo sapiens 35-39 24613679-9 2014 CPZ inhibited the induction of the HO-1 expression by rHx-bound heme. Heme 64-68 heme oxygenase 1 Homo sapiens 35-39 24613679-10 2014 CONCLUSION: rHx-bound heme was internalized into the cells via endocytosis, resulting in HO-1 expression and inactivation of Bach1. rhx 12-15 heme oxygenase 1 Homo sapiens 89-93 24613679-10 2014 CONCLUSION: rHx-bound heme was internalized into the cells via endocytosis, resulting in HO-1 expression and inactivation of Bach1. Heme 22-26 heme oxygenase 1 Homo sapiens 89-93 24613679-11 2014 GENERAL SIGNIFICANCE: The Bach1-dependent repression of the HO-1 expression is under the control of the Hx-dependent uptake of extracellular heme. Heme 141-145 heme oxygenase 1 Homo sapiens 60-64 24613679-2 2014 Heme also induces the expression of genes such as heme oxygenase-1 (HO-1) by inactivating the transcription repressor Bach1 through direct binding. Heme 0-4 heme oxygenase 1 Homo sapiens 50-66 24613679-2 2014 Heme also induces the expression of genes such as heme oxygenase-1 (HO-1) by inactivating the transcription repressor Bach1 through direct binding. Heme 0-4 heme oxygenase 1 Homo sapiens 68-72 24613679-3 2014 However, the source of heme for the regulation of the Bach1-HO-1 axis has been unclear. Heme 23-27 heme oxygenase 1 Homo sapiens 60-64 25022355-7 2014 Also, SR decreased Ca(2+) and ROS, mitochondrial membrane potential, dysfunction, and increased glutathione, NAD(P)H dehydrogenase and heme oxygenase-1. Strontium 6-8 heme oxygenase 1 Homo sapiens 135-151 24509340-2 2014 Colon and pancreas tumor cells have an increase in hemeoxygenase-1 (HO-1) activity, the endogenous enzyme responsible for carbon monoxide production. Carbon Monoxide 122-137 heme oxygenase 1 Homo sapiens 51-72 24879794-6 2014 Exogenous hemin induced Treg polarization in purified T cell/monocyte cocultures from healthy volunteers through the monocyte anti-inflammatory heme-degrading enzyme heme oxygenase-1. Hemin 10-15 heme oxygenase 1 Homo sapiens 166-182 24859911-12 2014 This is probably due to the enhancement of oxidative stress and cellular apoptosis induced by frequent glucose swings through the inhibition of Nrf2/HO-1 pathway. Glucose 103-110 heme oxygenase 1 Homo sapiens 149-153 24742817-6 2014 Increased O2(-) levels promote ERK phosphorylation (approx sixfold compared to control; P < 0.001) and downstream transcriptional increase of several genes: HMOX1 (P < 0.05), involved in the protection of chemical reactive species; MDR1 (P < 0.01), responsible for the efflux of xenobiotics in the cell; and CD83 (P < 0.05), a DC maturation marker. Superoxides 10-12 heme oxygenase 1 Homo sapiens 160-165 24799610-4 2014 Individuals with the long allele of a guanine-thymine (GTn) microsatellite repeat located in the promoter region of HMOX1 have a higher risk of cardiometabolic diseases compared with those with the short allele. guanine-thymine 38-53 heme oxygenase 1 Homo sapiens 116-121 24799610-4 2014 Individuals with the long allele of a guanine-thymine (GTn) microsatellite repeat located in the promoter region of HMOX1 have a higher risk of cardiometabolic diseases compared with those with the short allele. Nitroglycerin 55-58 heme oxygenase 1 Homo sapiens 116-121 24670329-5 2014 We and others have demonstrated that heme oxygenase-1 can modulate production of reactive oxygen species, both in vivo and in vitro, from NADPH oxidase, a major enzymatic source of reactive oxygen species generation in the vascular wall. Reactive Oxygen Species 81-104 heme oxygenase 1 Homo sapiens 37-53 24670329-5 2014 We and others have demonstrated that heme oxygenase-1 can modulate production of reactive oxygen species, both in vivo and in vitro, from NADPH oxidase, a major enzymatic source of reactive oxygen species generation in the vascular wall. Reactive Oxygen Species 181-204 heme oxygenase 1 Homo sapiens 37-53 24879794-6 2014 Exogenous hemin induced Treg polarization in purified T cell/monocyte cocultures from healthy volunteers through the monocyte anti-inflammatory heme-degrading enzyme heme oxygenase-1. Heme 144-148 heme oxygenase 1 Homo sapiens 166-182 26104642-6 2014 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 72-87 heme oxygenase 1 Homo sapiens 29-45 24673378-0 2014 Guanosine protects C6 astroglial cells against azide-induced oxidative damage: a putative role of heme oxygenase 1. Guanosine 0-9 heme oxygenase 1 Homo sapiens 98-114 24673378-8 2014 Guanosine strongly prevented these effects, protecting glial cells against azide-induced cytotoxicity and modulating glial, oxidative and inflammatory responses through the activation of the heme oxygenase 1 pathway. Guanosine 0-9 heme oxygenase 1 Homo sapiens 191-207 24673378-12 2014 Guanosine prevented these effects through the HO1 signaling pathway, thus our findings support the antioxidant effects of guanosine. Guanosine 0-9 heme oxygenase 1 Homo sapiens 46-49 24673378-12 2014 Guanosine prevented these effects through the HO1 signaling pathway, thus our findings support the antioxidant effects of guanosine. Guanosine 122-131 heme oxygenase 1 Homo sapiens 46-49 24788892-5 2014 Therefore, the present study assessed the inhibitory effect of PEITC on K562 cells and whether HO-1 facilitated cell apoptosis and ROS generation. Reactive Oxygen Species 131-134 heme oxygenase 1 Homo sapiens 95-99 24788892-8 2014 The results of the present study suggest that PEITC may be a potential anti-tumor compound for CML therapy, and that HO-1 has a critical function in PEITC-induced apoptosis and ROS generation. phenethyl isothiocyanate 149-154 heme oxygenase 1 Homo sapiens 117-121 24788892-8 2014 The results of the present study suggest that PEITC may be a potential anti-tumor compound for CML therapy, and that HO-1 has a critical function in PEITC-induced apoptosis and ROS generation. Reactive Oxygen Species 177-180 heme oxygenase 1 Homo sapiens 117-121 24676542-0 2014 Heme oxygenase-1 silencing increases the sensitivity of human osteosarcoma MG63 cells to arsenic trioxide. Arsenic Trioxide 89-105 heme oxygenase 1 Homo sapiens 0-16 24676542-2 2014 Our previous study regarding the effects of ATO on mesenchymal-derived human osteosarcoma MG63 cells showed that heme oxygenase-1 (HO-1) was strongly induced upon treatment with ATO. Arsenic Trioxide 44-47 heme oxygenase 1 Homo sapiens 113-129 24676542-2 2014 Our previous study regarding the effects of ATO on mesenchymal-derived human osteosarcoma MG63 cells showed that heme oxygenase-1 (HO-1) was strongly induced upon treatment with ATO. Arsenic Trioxide 44-47 heme oxygenase 1 Homo sapiens 131-135 24676542-2 2014 Our previous study regarding the effects of ATO on mesenchymal-derived human osteosarcoma MG63 cells showed that heme oxygenase-1 (HO-1) was strongly induced upon treatment with ATO. Arsenic Trioxide 178-181 heme oxygenase 1 Homo sapiens 113-129 24676542-2 2014 Our previous study regarding the effects of ATO on mesenchymal-derived human osteosarcoma MG63 cells showed that heme oxygenase-1 (HO-1) was strongly induced upon treatment with ATO. Arsenic Trioxide 178-181 heme oxygenase 1 Homo sapiens 131-135 24676542-3 2014 The present study sought to investigate the effect of silencing HO-1 on the sensitivity of osteosarcoma cells to ATO to determine the potential for therapeutic applications. Arsenic Trioxide 113-116 heme oxygenase 1 Homo sapiens 64-68 24676542-6 2014 shRNA-mediated interference prevented the induction of HO-1, increased cell death, and increased intracellular ROS levels in MG63 cells upon treatment with ATO. Arsenic Trioxide 156-159 heme oxygenase 1 Homo sapiens 55-59 24676542-7 2014 Silencing HO-1 increased the susceptibility of MG63 cells to the chemotherapeutic drug ATO by enhancing intracellular accumulation of ROS. Reactive Oxygen Species 134-137 heme oxygenase 1 Homo sapiens 10-14 24676542-8 2014 Our results suggest that the inhibition of HO-1 could improve the outcome of osteosarcoma treated with ATO. Arsenic Trioxide 103-106 heme oxygenase 1 Homo sapiens 43-47 26104642-6 2014 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 29-45 26104642-6 2014 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 47-51 26104642-6 2014 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 72-87 heme oxygenase 1 Homo sapiens 47-51 24530428-9 2014 In the case of DP41, the production of O2(-) radicals was followed by an up-regulation of Nrf2, HO-1, phospho-eIF2alpha and ATF4. o2(-) radicals 39-53 heme oxygenase 1 Homo sapiens 96-100 24792517-4 2014 Although hemin was able to induce HO-1 in a dose dependent manner, a specific HO-1 inhibitor, Sn-PPIX, was unable to interfere with the effect of hemin on Caco-2 cells. Hemin 9-14 heme oxygenase 1 Homo sapiens 34-38 24792517-4 2014 Although hemin was able to induce HO-1 in a dose dependent manner, a specific HO-1 inhibitor, Sn-PPIX, was unable to interfere with the effect of hemin on Caco-2 cells. sn-ppix 94-101 heme oxygenase 1 Homo sapiens 78-82 24958774-4 2014 In a stable isotope labeling by amino acids in cell culture-based proteomics screen, we identified HO-1 (heme oxygenase-1), the rate-limiting enzyme in the degradation of heme to biliverdin, as a novel SPP substrate. Heme 105-109 heme oxygenase 1 Homo sapiens 99-103 24958774-4 2014 In a stable isotope labeling by amino acids in cell culture-based proteomics screen, we identified HO-1 (heme oxygenase-1), the rate-limiting enzyme in the degradation of heme to biliverdin, as a novel SPP substrate. Biliverdine 179-189 heme oxygenase 1 Homo sapiens 99-103 24958774-4 2014 In a stable isotope labeling by amino acids in cell culture-based proteomics screen, we identified HO-1 (heme oxygenase-1), the rate-limiting enzyme in the degradation of heme to biliverdin, as a novel SPP substrate. Biliverdine 179-189 heme oxygenase 1 Homo sapiens 105-121 24927633-0 2014 Upregulation of heme oxygenase-1 in colorectal cancer patients with increased circulation carbon monoxide levels, potentially affects chemotherapeutic sensitivity. Carbon Monoxide 90-105 heme oxygenase 1 Homo sapiens 16-32 24927633-1 2014 BACKGROUND: Heme oxygenase-1 (HO-1) and its major product carbon monoxide (CO) are known to be involved in the development and progression of many tumors. Carbon Monoxide 58-73 heme oxygenase 1 Homo sapiens 12-28 24927633-1 2014 BACKGROUND: Heme oxygenase-1 (HO-1) and its major product carbon monoxide (CO) are known to be involved in the development and progression of many tumors. Carbon Monoxide 58-73 heme oxygenase 1 Homo sapiens 30-34 24927633-1 2014 BACKGROUND: Heme oxygenase-1 (HO-1) and its major product carbon monoxide (CO) are known to be involved in the development and progression of many tumors. Carbon Monoxide 75-77 heme oxygenase 1 Homo sapiens 12-28 24927633-1 2014 BACKGROUND: Heme oxygenase-1 (HO-1) and its major product carbon monoxide (CO) are known to be involved in the development and progression of many tumors. Carbon Monoxide 75-77 heme oxygenase 1 Homo sapiens 30-34 24927633-9 2014 In parallel with HO-1 expression, circulation CO levels in CRC patients also significantly accelerated. Carbon Monoxide 46-48 heme oxygenase 1 Homo sapiens 17-21 24927633-10 2014 Moreover, HO-1 expression/induction also related to the chemosensitivity of colon cells; HO inhibitor zinc protoporphyrin significantly increased cytotoxicities of THP (i.e., 2.6 - 5.3 folds compared to cells without zinc protoporphyrin treatment). zinc protoporphyrin 102-121 heme oxygenase 1 Homo sapiens 10-14 24927633-10 2014 Moreover, HO-1 expression/induction also related to the chemosensitivity of colon cells; HO inhibitor zinc protoporphyrin significantly increased cytotoxicities of THP (i.e., 2.6 - 5.3 folds compared to cells without zinc protoporphyrin treatment). pirarubicin 164-167 heme oxygenase 1 Homo sapiens 10-14 24927633-10 2014 Moreover, HO-1 expression/induction also related to the chemosensitivity of colon cells; HO inhibitor zinc protoporphyrin significantly increased cytotoxicities of THP (i.e., 2.6 - 5.3 folds compared to cells without zinc protoporphyrin treatment). zinc protoporphyrin 217-236 heme oxygenase 1 Homo sapiens 10-14 24041027-2 2014 We sought to determine the role of NO, heme oxygenase-1 (HO-1), and its reaction product (carbon monoxide [CO]) in the induction of mitochondrial biogenesis by the natural antioxidant resveratrol. Resveratrol 184-195 heme oxygenase 1 Homo sapiens 39-55 24041027-2 2014 We sought to determine the role of NO, heme oxygenase-1 (HO-1), and its reaction product (carbon monoxide [CO]) in the induction of mitochondrial biogenesis by the natural antioxidant resveratrol. Resveratrol 184-195 heme oxygenase 1 Homo sapiens 57-61 24041027-6 2014 Cobalt protoporphyrin (CoPP)-IX, an HO-1 inducing agent, stimulated mitochondrial biogenesis in HepG2 cells, which could be reversed by the CO scavenger hemoglobin. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 36-40 24041027-6 2014 Cobalt protoporphyrin (CoPP)-IX, an HO-1 inducing agent, stimulated mitochondrial biogenesis in HepG2 cells, which could be reversed by the CO scavenger hemoglobin. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 36-40 24041027-9 2014 The natural antioxidant resveratrol induced mitochondrial biogenesis in HepG2 cells, in a manner dependent on NO biosynthesis, cGMP synthesis, Nrf2-dependent HO-1 activation, and endogenous CO production. Resveratrol 24-35 heme oxygenase 1 Homo sapiens 158-162 24959672-6 2014 In addition, urate markedly up-regulated the transcription and protein expression of gamma-glutamate-cysteine ligase catalytic subunit (gamma-GCLC) and heme oxygenase-1 (HO-1), both of which are controlled by Nrf2 activity. Uric Acid 13-18 heme oxygenase 1 Homo sapiens 152-168 24768191-2 2014 Niacin directly activates both GPR-109A in leukocytes and the heme oxygenase-1 pathway, promoting strong anti-inflammatory and antioxidative effects, as well as induces immediate production of prostaglandin D2, leading to endothelial vasodilation. Niacin 0-6 heme oxygenase 1 Homo sapiens 62-78 24458266-7 2014 HO-1 concentrations also positively correlated with random plasma glucose, circulating glycosylated hemoglobin, and low-density lipoprotein levels. Glucose 66-73 heme oxygenase 1 Homo sapiens 0-4 24458266-8 2014 Moreover, patients with coincident TB and T2DM exhibited increased plasma levels of TIMP-4 and elevated peripheral blood neutrophil counts, which, when considered together with HO-1, resulted in increased power to discriminate individuals with active TB with and without T2DM. Terbium 35-37 heme oxygenase 1 Homo sapiens 177-181 24610348-5 2014 In addition, DHF induced the activation of extracellular signal-regulated kinase (ERK), while U0126 (a specific pharmacological inhibitor of ERK kinase) abrogated DHF-activated Nrf2 and HO-1 expression. dhf 163-166 heme oxygenase 1 Homo sapiens 186-190 24943970-0 2014 The relationship between anticancer effect of metformin and the transcriptional regulation of certain genes (CHOP, CAV-1, HO-1, SGK-1 and Par-4) on MCF-7 cell line. Metformin 46-55 heme oxygenase 1 Homo sapiens 122-126 24610348-0 2014 Effect of 7, 8-dihydroxyflavone on the up-regulation of Nrf2-mediated heme oxygenase-1 expression in hamster lung fibroblasts. 6,7-dihydroxyflavone 10-31 heme oxygenase 1 Homo sapiens 70-86 24442713-0 2014 Angiotensin II-derived reactive oxygen species promote angiogenesis in human late endothelial progenitor cells through heme oxygenase-1 via ERK1/2 and AKT/PI3K pathways. Reactive Oxygen Species 23-46 heme oxygenase 1 Homo sapiens 119-135 24442713-4 2014 Ang II-derived ROS could also upregulate heme oxygenase-1 (HO-1) expression, and treating late EPCs with HO-1 small interfering RNA or heme oxygenase inhibitor (HO inhibitor) could inhibit Ang II-induced tube formation and increase ROS level and apoptosis rate. Reactive Oxygen Species 15-18 heme oxygenase 1 Homo sapiens 41-57 24442713-4 2014 Ang II-derived ROS could also upregulate heme oxygenase-1 (HO-1) expression, and treating late EPCs with HO-1 small interfering RNA or heme oxygenase inhibitor (HO inhibitor) could inhibit Ang II-induced tube formation and increase ROS level and apoptosis rate. Reactive Oxygen Species 15-18 heme oxygenase 1 Homo sapiens 59-63 24442713-4 2014 Ang II-derived ROS could also upregulate heme oxygenase-1 (HO-1) expression, and treating late EPCs with HO-1 small interfering RNA or heme oxygenase inhibitor (HO inhibitor) could inhibit Ang II-induced tube formation and increase ROS level and apoptosis rate. Reactive Oxygen Species 232-235 heme oxygenase 1 Homo sapiens 105-109 24442713-5 2014 In addition, PD98059 and LY294002 pretreatment attenuated Ang II-induced HO-1 expression. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 13-20 heme oxygenase 1 Homo sapiens 73-77 24442713-5 2014 In addition, PD98059 and LY294002 pretreatment attenuated Ang II-induced HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 25-33 heme oxygenase 1 Homo sapiens 73-77 24442713-6 2014 Accordingly, Ang II-derived ROS could promote angiogenesis in late EPCs by inducing HO-1 expression via ERK1/2 and AKT/PI3K pathways, and we believe HO-1 might be a promising intervention target in EPCs due to its potent proangiogenic, antioxidant, and antiapoptosis potentials. Reactive Oxygen Species 28-31 heme oxygenase 1 Homo sapiens 84-88 24442713-6 2014 Accordingly, Ang II-derived ROS could promote angiogenesis in late EPCs by inducing HO-1 expression via ERK1/2 and AKT/PI3K pathways, and we believe HO-1 might be a promising intervention target in EPCs due to its potent proangiogenic, antioxidant, and antiapoptosis potentials. Reactive Oxygen Species 28-31 heme oxygenase 1 Homo sapiens 149-153 24610348-1 2014 The cytoprotective mechanism of 7, 8-dihydroxyflavone (DHF) against oxidative stress-induced cell damage with respect to its stimulatory effect on the expression of heme oxygenase-1 (HO-1), a potent antioxidant enzyme, was investigated in the present study. 6,7-dihydroxyflavone 32-53 heme oxygenase 1 Homo sapiens 165-181 24610348-1 2014 The cytoprotective mechanism of 7, 8-dihydroxyflavone (DHF) against oxidative stress-induced cell damage with respect to its stimulatory effect on the expression of heme oxygenase-1 (HO-1), a potent antioxidant enzyme, was investigated in the present study. 6,7-dihydroxyflavone 32-53 heme oxygenase 1 Homo sapiens 183-187 24610348-1 2014 The cytoprotective mechanism of 7, 8-dihydroxyflavone (DHF) against oxidative stress-induced cell damage with respect to its stimulatory effect on the expression of heme oxygenase-1 (HO-1), a potent antioxidant enzyme, was investigated in the present study. dhf 55-58 heme oxygenase 1 Homo sapiens 165-181 24610348-1 2014 The cytoprotective mechanism of 7, 8-dihydroxyflavone (DHF) against oxidative stress-induced cell damage with respect to its stimulatory effect on the expression of heme oxygenase-1 (HO-1), a potent antioxidant enzyme, was investigated in the present study. dhf 55-58 heme oxygenase 1 Homo sapiens 183-187 24610348-2 2014 Up-regulation of HO-1 expression by DHF was both dose and time dependent in lung fibroblast V79-4 cells. dhf 36-39 heme oxygenase 1 Homo sapiens 17-21 24610348-3 2014 DHF also increased the protein expression level of the transcription factor nuclear factor erythroid-2-related factor 2 (Nrf2), and induced the translocation of Nrf2 from the cytosol into the nucleus, leading to elevated HO-1 expression. dhf 0-3 heme oxygenase 1 Homo sapiens 221-225 24610348-4 2014 The siNrf2 RNA-transfection attenuated HO-1 expression induced by DHF treatment. dhf 66-69 heme oxygenase 1 Homo sapiens 39-43 24610348-5 2014 In addition, DHF induced the activation of extracellular signal-regulated kinase (ERK), while U0126 (a specific pharmacological inhibitor of ERK kinase) abrogated DHF-activated Nrf2 and HO-1 expression. U 0126 94-99 heme oxygenase 1 Homo sapiens 186-190 24610348-6 2014 This suggests that DHF increased the levels of Nrf2 and HO-1 via ERK-dependent pathways. dhf 19-22 heme oxygenase 1 Homo sapiens 56-60 24610348-8 2014 Taken together, these results demonstrate that DHF protected cells from oxidative stress via the activation of an ERK/Nrf2/HO-1 signaling pathway. dhf 47-50 heme oxygenase 1 Homo sapiens 123-127 24415199-5 2014 Our study demonstrated that DMSO pretreatment showed a cytoprotective effect against H2 O2 -induced oxidative damage (impaired cell viability, increased apopototic cells rate and caspase-3 level, and increased release of LDH and CK) and this process is partially mediated by HO-1 upregulation. Dimethyl Sulfoxide 28-32 heme oxygenase 1 Homo sapiens 275-279 24415199-0 2014 Dimethyl sulfoxide attenuates hydrogen peroxide-induced injury in cardiomyocytes via heme oxygenase-1. Dimethyl Sulfoxide 0-18 heme oxygenase 1 Homo sapiens 85-101 24415199-5 2014 Our study demonstrated that DMSO pretreatment showed a cytoprotective effect against H2 O2 -induced oxidative damage (impaired cell viability, increased apopototic cells rate and caspase-3 level, and increased release of LDH and CK) and this process is partially mediated by HO-1 upregulation. Hydrogen Peroxide 85-90 heme oxygenase 1 Homo sapiens 275-279 24415199-0 2014 Dimethyl sulfoxide attenuates hydrogen peroxide-induced injury in cardiomyocytes via heme oxygenase-1. Hydrogen Peroxide 30-47 heme oxygenase 1 Homo sapiens 85-101 24415199-6 2014 Furthermore, our data showed that the activation of p38 MAPK and Nrf2 translocation are involved in the HO-1 upregulation induced by DMSO. Dimethyl Sulfoxide 133-137 heme oxygenase 1 Homo sapiens 104-108 24415199-2 2014 Our former study showed that DMSO is able to induce heme oxygenase-1 (HO-1) expression in endothelial cells, which is a potent antioxidant enzyme. Dimethyl Sulfoxide 29-33 heme oxygenase 1 Homo sapiens 52-68 24415199-7 2014 This study reports for the first time that the cytoprotective effect of DMSO in cardiomyocytes is partially mediated by HO-1, which may further explain the mechanisms by which DMSO exerts cardioprotection on H2 O2 injury. Dimethyl Sulfoxide 72-76 heme oxygenase 1 Homo sapiens 120-124 24415199-2 2014 Our former study showed that DMSO is able to induce heme oxygenase-1 (HO-1) expression in endothelial cells, which is a potent antioxidant enzyme. Dimethyl Sulfoxide 29-33 heme oxygenase 1 Homo sapiens 70-74 24415199-3 2014 In this study, we hypothesized that the antioxidant effects of DMSO in cardiomyocytes are mediated or partially mediated by increased HO-1 expression. Dimethyl Sulfoxide 63-67 heme oxygenase 1 Homo sapiens 134-138 24415199-7 2014 This study reports for the first time that the cytoprotective effect of DMSO in cardiomyocytes is partially mediated by HO-1, which may further explain the mechanisms by which DMSO exerts cardioprotection on H2 O2 injury. Dimethyl Sulfoxide 176-180 heme oxygenase 1 Homo sapiens 120-124 24415199-7 2014 This study reports for the first time that the cytoprotective effect of DMSO in cardiomyocytes is partially mediated by HO-1, which may further explain the mechanisms by which DMSO exerts cardioprotection on H2 O2 injury. Hydrogen Peroxide 208-213 heme oxygenase 1 Homo sapiens 120-124 24500083-8 2014 Moreover, a regulatory role of ROS for HO-1 regulation in these cells is demonstrated by studies with the antioxidant N-acetylcysteine and exogenous hydrogenperoxide. Reactive Oxygen Species 31-34 heme oxygenase 1 Homo sapiens 39-43 24413982-8 2014 Treatment with resveratrol and genipin up-regulated NQO1 and HMOX1 expression and reduced oxidative stress in patients" lymphoblastoid cells. Resveratrol 15-26 heme oxygenase 1 Homo sapiens 61-66 24413982-8 2014 Treatment with resveratrol and genipin up-regulated NQO1 and HMOX1 expression and reduced oxidative stress in patients" lymphoblastoid cells. genipin 31-38 heme oxygenase 1 Homo sapiens 61-66 24977002-1 2014 BACKGROUND: There is a reverse relationship between serum bilirubin level and incidence of stroke, heme oxygenase-1 (HO-1) can catalyze heme into bilirubin, it is unknown the association of HO-1 level with risk of stroke. Heme 99-103 heme oxygenase 1 Homo sapiens 117-121 24977002-1 2014 BACKGROUND: There is a reverse relationship between serum bilirubin level and incidence of stroke, heme oxygenase-1 (HO-1) can catalyze heme into bilirubin, it is unknown the association of HO-1 level with risk of stroke. Bilirubin 146-155 heme oxygenase 1 Homo sapiens 99-115 24977002-1 2014 BACKGROUND: There is a reverse relationship between serum bilirubin level and incidence of stroke, heme oxygenase-1 (HO-1) can catalyze heme into bilirubin, it is unknown the association of HO-1 level with risk of stroke. Bilirubin 146-155 heme oxygenase 1 Homo sapiens 117-121 24412540-6 2014 Resveratrol decreases intracellular ROS production, most likely by mechanisms involving NMDA, AMPA/KA, intracellular Ca(2+) and the heme oxygenase 1 (HO1) pathway, and prevents mitochondrial dysfunction and impairments in Na(+)K(+)-ATPase and GS activity after glutamate activation. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 132-148 24412540-6 2014 Resveratrol decreases intracellular ROS production, most likely by mechanisms involving NMDA, AMPA/KA, intracellular Ca(2+) and the heme oxygenase 1 (HO1) pathway, and prevents mitochondrial dysfunction and impairments in Na(+)K(+)-ATPase and GS activity after glutamate activation. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 150-153 24412540-6 2014 Resveratrol decreases intracellular ROS production, most likely by mechanisms involving NMDA, AMPA/KA, intracellular Ca(2+) and the heme oxygenase 1 (HO1) pathway, and prevents mitochondrial dysfunction and impairments in Na(+)K(+)-ATPase and GS activity after glutamate activation. Reactive Oxygen Species 36-39 heme oxygenase 1 Homo sapiens 132-148 24736402-3 2014 To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). cobaltiprotoporphyrin 63-84 heme oxygenase 1 Homo sapiens 139-155 24736402-3 2014 To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). cobaltiprotoporphyrin 63-84 heme oxygenase 1 Homo sapiens 157-161 24736402-3 2014 To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). cobaltiprotoporphyrin 86-90 heme oxygenase 1 Homo sapiens 139-155 24736402-3 2014 To improve early graft survival, hESC-CMs were pretreated with cobalt protoporphyrin (CoPP), a transcriptional activator of cytoprotective heme oxygenase-1 (HO-1). cobaltiprotoporphyrin 86-90 heme oxygenase 1 Homo sapiens 157-161 24736402-5 2014 Compared with PBS-pretreated cells, CoPP-pretreated hESC-CM preparations exhibited higher levels of HO-1 expression, Akt phosphorylation, and vascular endothelial growth factor production, with reduced apoptosis, and a 30% decrease in intracellular reactive oxygen species. cobaltiprotoporphyrin 36-40 heme oxygenase 1 Homo sapiens 100-104 24500083-8 2014 Moreover, a regulatory role of ROS for HO-1 regulation in these cells is demonstrated by studies with the antioxidant N-acetylcysteine and exogenous hydrogenperoxide. Acetylcysteine 118-134 heme oxygenase 1 Homo sapiens 39-43 24500083-8 2014 Moreover, a regulatory role of ROS for HO-1 regulation in these cells is demonstrated by studies with the antioxidant N-acetylcysteine and exogenous hydrogenperoxide. Hydrogen Peroxide 149-165 heme oxygenase 1 Homo sapiens 39-43 24801208-0 2014 Matrix conditions and KLF2-dependent induction of heme oxygenase-1 modulate inhibition of HCV replication by fluvastatin. Fluvastatin 109-120 heme oxygenase 1 Homo sapiens 50-66 24622883-5 2014 DFO and PHEN downregulated Grp78, Grp94, and MRP1 expressions and upregulated CHOP and HO-1 expressions. Deferoxamine 0-3 heme oxygenase 1 Homo sapiens 87-91 24622883-8 2014 We determined that the reduction of increased Grp78, Grp94, HO-1, and MRP1 expressions, which appears to inhibit the chemotherapeutic effect of TSA, through the combination with DFO or PHEN will contribute to the anticancer effect. trichostatin A 144-147 heme oxygenase 1 Homo sapiens 60-64 24622883-8 2014 We determined that the reduction of increased Grp78, Grp94, HO-1, and MRP1 expressions, which appears to inhibit the chemotherapeutic effect of TSA, through the combination with DFO or PHEN will contribute to the anticancer effect. Deferoxamine 178-181 heme oxygenase 1 Homo sapiens 60-64 24862399-8 2014 The Pearson correlation analysis showed that HO-1, HO-2, and HO activity were negatively associated with waist circumference (P<.05), body mass index (P<.05), triglycerides (P<.05) and positively with HDL-C (P<.05). Triglycerides 165-178 heme oxygenase 1 Homo sapiens 45-49 24862399-13 2014 CONCLUSION: Alteration of serum triglycerides and HDL-C significantly impairs HO-1 and HO-2 levels in benign prostatic hyperplasia patients. Triglycerides 32-45 heme oxygenase 1 Homo sapiens 78-82 24857917-0 2014 The cytoprotective effect of sulfuretin against tert-butyl hydroperoxide-induced hepatotoxicity through Nrf2/ARE and JNK/ERK MAPK-mediated heme oxygenase-1 expression. sulfuretin 29-39 heme oxygenase 1 Homo sapiens 139-155 24857917-0 2014 The cytoprotective effect of sulfuretin against tert-butyl hydroperoxide-induced hepatotoxicity through Nrf2/ARE and JNK/ERK MAPK-mediated heme oxygenase-1 expression. tert-Butylhydroperoxide 48-72 heme oxygenase 1 Homo sapiens 139-155 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. indoleacetic acid 64-67 heme oxygenase 1 Homo sapiens 171-187 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. indoleacetic acid 64-67 heme oxygenase 1 Homo sapiens 189-193 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. Hydrogen Sulfide 102-106 heme oxygenase 1 Homo sapiens 171-187 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. Hydrogen Sulfide 102-106 heme oxygenase 1 Homo sapiens 189-193 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. Carbon Monoxide 111-113 heme oxygenase 1 Homo sapiens 171-187 24832131-5 2014 Interestingly, during the initiation of lateral root primordia, IAA is a potent inducer of endogenous H 2S and CO, which is produced by L-cysteine desulfhydrase (LCD) and heme oxygenase-1 (HO-1), respectively. Carbon Monoxide 111-113 heme oxygenase 1 Homo sapiens 189-193 24622883-8 2014 We determined that the reduction of increased Grp78, Grp94, HO-1, and MRP1 expressions, which appears to inhibit the chemotherapeutic effect of TSA, through the combination with DFO or PHEN will contribute to the anticancer effect. Phenanthrolines 185-189 heme oxygenase 1 Homo sapiens 60-64 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Curcumin 12-20 heme oxygenase 1 Homo sapiens 47-63 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Curcumin 12-20 heme oxygenase 1 Homo sapiens 65-69 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Hemin 26-31 heme oxygenase 1 Homo sapiens 47-63 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Hemin 26-31 heme oxygenase 1 Homo sapiens 65-69 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Porphyrins 123-132 heme oxygenase 1 Homo sapiens 47-63 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Porphyrins 123-132 heme oxygenase 1 Homo sapiens 65-69 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Chlorophyll 137-148 heme oxygenase 1 Homo sapiens 47-63 24830678-7 2014 We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5x10-5). Chlorophyll 137-148 heme oxygenase 1 Homo sapiens 65-69 24830678-10 2014 Selective inhibition of HO-1 completely blocked the action of hemin but not that of curcumin, suggesting simultaneous multi-pathway intervention by curcumin. Curcumin 148-156 heme oxygenase 1 Homo sapiens 24-28 24819505-4 2014 Overexpression of Nrf2 or HO-1 resulted in upregulation of TP in NCI-H292 cells, an effect mimicked by treatment with an antioxidant N-acetylcysteine and partially reversed by HO-1 knockdown. Acetylcysteine 133-149 heme oxygenase 1 Homo sapiens 26-30 24553299-8 2014 Using an in vitro model, we found that HO-1 induction, using cobalt protoporphyrin, stimulates cytotrophoblast beta-ENaC expression by 1.5- and 1.8-fold (10 and 50 muM). cobaltiprotoporphyrin 61-82 heme oxygenase 1 Homo sapiens 39-43 24413553-2 2014 In addition to increased thrombin generation via tumor release of tissue factor-bearing microparticles and hyperfibrinogenemia, brain tumors and surrounding normal brain likely generate endogenous carbon monoxide (CO) via the hemeoxygenase-1 (HO-1) system. Carbon Monoxide 197-212 heme oxygenase 1 Homo sapiens 226-247 24413553-2 2014 In addition to increased thrombin generation via tumor release of tissue factor-bearing microparticles and hyperfibrinogenemia, brain tumors and surrounding normal brain likely generate endogenous carbon monoxide (CO) via the hemeoxygenase-1 (HO-1) system. Carbon Monoxide 214-216 heme oxygenase 1 Homo sapiens 226-247 24180257-7 2014 Further, heme is an important component of a number of metabolic enzymes, and, therefore, HO-1 plays an important role in the modulation of cellular bioenergetics. Heme 9-13 heme oxygenase 1 Homo sapiens 90-94 24743738-8 2014 The siRNA- or shRNA-mediated knockdown of Nrf2 or HO-1 significantly suppressed cancer cell viability and tumor growth in vitro and in vivo, resulting in enhanced 5-FU sensitivity. Fluorouracil 163-167 heme oxygenase 1 Homo sapiens 50-54 24743738-12 2014 These results suggested that the mechanism underlying the acquisition of 5-FU resistance in CRC involves the upregulation of Nrf2 and HO-1 expression via epigenetic modifications of DNA demethylation. Fluorouracil 73-77 heme oxygenase 1 Homo sapiens 134-138 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Heme 47-51 heme oxygenase 1 Homo sapiens 14-30 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Heme 47-51 heme oxygenase 1 Homo sapiens 32-36 24583029-3 2014 Hemin, a commercial preparation of heme, is used as an iron donor or heme oxygenase 1 (HO-1) inducer, and has been shown to provide antiviral activity in many studies. Hemin 0-5 heme oxygenase 1 Homo sapiens 69-85 24583029-3 2014 Hemin, a commercial preparation of heme, is used as an iron donor or heme oxygenase 1 (HO-1) inducer, and has been shown to provide antiviral activity in many studies. Hemin 0-5 heme oxygenase 1 Homo sapiens 87-91 24583029-7 2014 In addition, after treatment with Protoporphyrin IX zinc (II) (ZnPP) or Sn (IV) Protoporphyrin IX dichloride (SnPP), inhibitors of HO-1, the inhibition of viral replication by hemin was partially reversed. protoporphyrin ix dichloride 80-108 heme oxygenase 1 Homo sapiens 131-135 24553817-0 2014 Maslinic acid protects vascular smooth muscle cells from oxidative stress through Akt/Nrf2/HO-1 pathway. maslinic acid 0-13 heme oxygenase 1 Homo sapiens 91-95 24553817-5 2014 Wortmannin suppression of Akt was able to abolish HO-1 upregulation in VSMCs, suggesting the requirement of Akt activation for MA effect on HO-1. Wortmannin 0-10 heme oxygenase 1 Homo sapiens 50-54 24553817-5 2014 Wortmannin suppression of Akt was able to abolish HO-1 upregulation in VSMCs, suggesting the requirement of Akt activation for MA effect on HO-1. Wortmannin 0-10 heme oxygenase 1 Homo sapiens 140-144 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Biliverdine 55-65 heme oxygenase 1 Homo sapiens 14-30 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Biliverdine 55-65 heme oxygenase 1 Homo sapiens 32-36 24180287-7 2014 Moreover, the potential for heme catabolism end products, such as carbon monoxide, to amplify the HMOX1 stress response should be considered. Carbon Monoxide 66-81 heme oxygenase 1 Homo sapiens 98-103 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Carbon Monoxide 67-82 heme oxygenase 1 Homo sapiens 14-30 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Carbon Monoxide 67-82 heme oxygenase 1 Homo sapiens 32-36 24180608-1 2014 SIGNIFICANCE: Heme oxygenase enzymes, which exist as constitutive (HO-2) and inducible (HO-1) isoforms, degrade heme to carbon monoxide (CO) and the bile pigment biliverdin. Heme 112-116 heme oxygenase 1 Homo sapiens 88-92 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. ammonium ferrous sulfate 88-95 heme oxygenase 1 Homo sapiens 14-30 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. ammonium ferrous sulfate 88-95 heme oxygenase 1 Homo sapiens 32-36 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Heme 144-148 heme oxygenase 1 Homo sapiens 14-30 24180608-1 2014 SIGNIFICANCE: Heme oxygenase enzymes, which exist as constitutive (HO-2) and inducible (HO-1) isoforms, degrade heme to carbon monoxide (CO) and the bile pigment biliverdin. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 88-92 24053682-1 2014 SIGNIFICANCE: Heme oxygenase-1 (HO-1) converts heme to biliverdin, carbon monoxide, and ferrous ions, but its cellular functions are far beyond heme metabolism. Heme 144-148 heme oxygenase 1 Homo sapiens 32-36 24053682-2 2014 HO-1 via heme removal and degradation products acts as a cytoprotective, anti-inflammatory, immunomodulatory, and proangiogenic protein, regulating also a cell cycle. Heme 9-13 heme oxygenase 1 Homo sapiens 0-4 24180608-1 2014 SIGNIFICANCE: Heme oxygenase enzymes, which exist as constitutive (HO-2) and inducible (HO-1) isoforms, degrade heme to carbon monoxide (CO) and the bile pigment biliverdin. Carbon Monoxide 137-139 heme oxygenase 1 Homo sapiens 88-92 24180608-1 2014 SIGNIFICANCE: Heme oxygenase enzymes, which exist as constitutive (HO-2) and inducible (HO-1) isoforms, degrade heme to carbon monoxide (CO) and the bile pigment biliverdin. Biliverdine 162-172 heme oxygenase 1 Homo sapiens 88-92 24124891-5 2014 While heme catabolism by heme oxygenase-1 (HO-1) prevents programmed cell death, this cytoprotective effect requires the co-expression of ferritin H (heart/heavy) chain (FTH), which controls the pro-oxidant effect of labile Fe released from the protoporphyrin IX ring of heme. Heme 6-10 heme oxygenase 1 Homo sapiens 25-41 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Heme 90-94 heme oxygenase 1 Homo sapiens 31-35 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 100-115 heme oxygenase 1 Homo sapiens 31-35 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 117-119 heme oxygenase 1 Homo sapiens 31-35 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Iron 122-126 heme oxygenase 1 Homo sapiens 31-35 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Biliverdine 132-142 heme oxygenase 1 Homo sapiens 31-35 24131232-1 2014 SIGNIFICANCE: Heme oxygenases (HO-1 and HO-2) catalyze the degradation of the pro-oxidant heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Bilirubin 179-188 heme oxygenase 1 Homo sapiens 31-35 24131232-5 2014 Moreover, therapeutic delivery of HO-1 products CO, biliverdin, and bilirubin has been shown to have favorable effects, notably on endothelial cells and in animal models of vascular disease. Biliverdine 52-62 heme oxygenase 1 Homo sapiens 34-38 24131232-5 2014 Moreover, therapeutic delivery of HO-1 products CO, biliverdin, and bilirubin has been shown to have favorable effects, notably on endothelial cells and in animal models of vascular disease. Bilirubin 68-77 heme oxygenase 1 Homo sapiens 34-38 24341409-1 2014 Not only double, Janus face, but numerous appearances characterize heme oxygenase-1 (HO-1), an inducible enzyme which main role is to degrade heme. Heme 67-71 heme oxygenase 1 Homo sapiens 85-89 24147608-6 2014 The importance of HO-1 in MP is emphasized by their expression of specific receptors that primarily function to ingest heme-containing substrate and deliver it to HO-1. Heme 119-123 heme oxygenase 1 Homo sapiens 18-22 24413389-0 2014 Crucial role of heme oxygenase-1 in the sensitivity of acute myeloid leukemia cell line Kasumi-1 to ursolic acid. ursolic acid 100-112 heme oxygenase 1 Homo sapiens 16-32 24700716-2 2014 They focus on a key enzyme involved in heme catabolism, heme oxygenase 1 (HO-1), which, ironically, has been poorly investigated in erythroid cells, the largest pool of heme-containing cells. Heme 39-43 heme oxygenase 1 Homo sapiens 56-72 24700716-2 2014 They focus on a key enzyme involved in heme catabolism, heme oxygenase 1 (HO-1), which, ironically, has been poorly investigated in erythroid cells, the largest pool of heme-containing cells. Heme 39-43 heme oxygenase 1 Homo sapiens 74-78 24413389-4 2014 HO-1 in Kasumi-1 cells was upregulated by being treated with low-dose rather than high-dose UA. ursolic acid 92-94 heme oxygenase 1 Homo sapiens 0-4 24413389-5 2014 Inhibition of HO-1 by zinc protoporphyrin (ZnPP) IX sensitized Kasumi-1 cells to UA, and the apoptotic rate was close to that induced by Ara-C (P<0.01). zinc protoporphyrin 22-41 heme oxygenase 1 Homo sapiens 14-18 24413389-5 2014 Inhibition of HO-1 by zinc protoporphyrin (ZnPP) IX sensitized Kasumi-1 cells to UA, and the apoptotic rate was close to that induced by Ara-C (P<0.01). zinc protoporphyrin 43-47 heme oxygenase 1 Homo sapiens 14-18 24413389-5 2014 Inhibition of HO-1 by zinc protoporphyrin (ZnPP) IX sensitized Kasumi-1 cells to UA, and the apoptotic rate was close to that induced by Ara-C (P<0.01). ursolic acid 81-83 heme oxygenase 1 Homo sapiens 14-18 24413389-7 2014 After silencing HO-1 by siRNA transfection with lentivirus, the cells" proliferation induced by UA was increased as it was by Ara-C. ursolic acid 96-98 heme oxygenase 1 Homo sapiens 16-20 24413389-7 2014 After silencing HO-1 by siRNA transfection with lentivirus, the cells" proliferation induced by UA was increased as it was by Ara-C. Cytarabine 126-131 heme oxygenase 1 Homo sapiens 16-20 24413389-10 2014 AML-M2 can feasibly be treated by target-inhibiting HO-1 that enhances the antileukemia effects of UA in vitro and in vivo. ursolic acid 99-101 heme oxygenase 1 Homo sapiens 52-56 24503931-0 2014 7,8-Dihydroxyflavone protects human keratinocytes against oxidative stress-induced cell damage via the ERK and PI3K/Akt-mediated Nrf2/HO-1 signaling pathways. 6,7-dihydroxyflavone 0-20 heme oxygenase 1 Homo sapiens 134-138 24847330-3 2014 HO-1 degrades heme but increases in experimental models of AKI. Heme 14-18 heme oxygenase 1 Homo sapiens 0-4 24588654-6 2014 Moreover, treatment of NHEKs with Z-Ligustilide increased reactive oxygen species (ROS) and L-N-acetylcysteine (L-NAC, an antioxidant) attenuated Z-ligustilide-induced Nrf2 nuclear translocation and HO-1 expression. ligustilide 34-47 heme oxygenase 1 Homo sapiens 199-203 24588654-6 2014 Moreover, treatment of NHEKs with Z-Ligustilide increased reactive oxygen species (ROS) and L-N-acetylcysteine (L-NAC, an antioxidant) attenuated Z-ligustilide-induced Nrf2 nuclear translocation and HO-1 expression. Acetylcysteine 92-110 heme oxygenase 1 Homo sapiens 199-203 24588654-6 2014 Moreover, treatment of NHEKs with Z-Ligustilide increased reactive oxygen species (ROS) and L-N-acetylcysteine (L-NAC, an antioxidant) attenuated Z-ligustilide-induced Nrf2 nuclear translocation and HO-1 expression. Acetylcysteine 112-117 heme oxygenase 1 Homo sapiens 199-203 24588654-6 2014 Moreover, treatment of NHEKs with Z-Ligustilide increased reactive oxygen species (ROS) and L-N-acetylcysteine (L-NAC, an antioxidant) attenuated Z-ligustilide-induced Nrf2 nuclear translocation and HO-1 expression. ligustilide 146-159 heme oxygenase 1 Homo sapiens 199-203 24380881-0 2014 HO-1 up-regulation: a key point in high-risk neuroblastoma resistance to bortezomib. Bortezomib 73-83 heme oxygenase 1 Homo sapiens 0-4 24380881-3 2014 We have shown that MYCN amplified HTLA-230 cells were slightly sensitive to BTZ treatment, due to the activation of Nrf2 that led to an impressive up-regulation of HO-1. Bortezomib 76-79 heme oxygenase 1 Homo sapiens 164-168 24380881-5 2014 The inhibition of HO-1 activity obtained by Zinc (II) protoprophyrin IX (ZnPPIX) was able to significantly increase the pro-apoptotic effect of BTZ in a p53- and p21-independent way. zinc (ii) protoprophyrin ix 44-71 heme oxygenase 1 Homo sapiens 18-22 24380881-5 2014 The inhibition of HO-1 activity obtained by Zinc (II) protoprophyrin IX (ZnPPIX) was able to significantly increase the pro-apoptotic effect of BTZ in a p53- and p21-independent way. zinc protoporphyrin 73-79 heme oxygenase 1 Homo sapiens 18-22 24380881-5 2014 The inhibition of HO-1 activity obtained by Zinc (II) protoprophyrin IX (ZnPPIX) was able to significantly increase the pro-apoptotic effect of BTZ in a p53- and p21-independent way. Bortezomib 144-147 heme oxygenase 1 Homo sapiens 18-22 24486344-0 2014 Salvianolic acid A protects RPE cells against oxidative stress through activation of Nrf2/HO-1 signaling. salvianolic acid A 0-18 heme oxygenase 1 Homo sapiens 90-94 24486344-7 2014 Both Nrf2 and HO-1 were required for Sal A-mediated cytoprotective effect, as Nrf2/HO-1 inhibition abolished Sal A-induced beneficial effects against H2O2. salvianolic acid A 37-42 heme oxygenase 1 Homo sapiens 14-18 24486344-7 2014 Both Nrf2 and HO-1 were required for Sal A-mediated cytoprotective effect, as Nrf2/HO-1 inhibition abolished Sal A-induced beneficial effects against H2O2. salvianolic acid A 37-42 heme oxygenase 1 Homo sapiens 83-87 24486344-7 2014 Both Nrf2 and HO-1 were required for Sal A-mediated cytoprotective effect, as Nrf2/HO-1 inhibition abolished Sal A-induced beneficial effects against H2O2. salvianolic acid A 109-114 heme oxygenase 1 Homo sapiens 83-87 24486344-7 2014 Both Nrf2 and HO-1 were required for Sal A-mediated cytoprotective effect, as Nrf2/HO-1 inhibition abolished Sal A-induced beneficial effects against H2O2. Hydrogen Peroxide 150-154 heme oxygenase 1 Homo sapiens 83-87 24486344-8 2014 Meanwhile, the PI3K/Akt/mTORC1 chemical inhibitors not only suppressed Sal A-induced Nrf2/HO-1 activation, but also eliminated its cytoprotective effect in RPE cells. salvianolic acid A 71-76 heme oxygenase 1 Homo sapiens 90-94 24486344-9 2014 These observations suggest that Sal A activates the Nrf2/HO-1 axis in RPE cells and protects against oxidative stress via activation of Akt/mTORC1 signaling. salvianolic acid A 32-37 heme oxygenase 1 Homo sapiens 57-61 24503931-7 2014 Taken together, the results suggested that DHF activates ERK- and Akt-Nrf2 signaling cascades in HaCaT cells, leading to the upregulation of HO-1 and cytoprotection against oxidative stress. 6,7-dihydroxyflavone 43-46 heme oxygenase 1 Homo sapiens 141-145 24503931-1 2014 This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. 6,7-dihydroxyflavone 38-58 heme oxygenase 1 Homo sapiens 99-115 24503931-1 2014 This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. 6,7-dihydroxyflavone 38-58 heme oxygenase 1 Homo sapiens 117-121 24503931-1 2014 This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. 6,7-dihydroxyflavone 60-63 heme oxygenase 1 Homo sapiens 99-115 24503931-1 2014 This study investigated the effect of 7,8-dihydroxyflavone (DHF) on the expression and activity of heme oxygenase-1 (HO-1), an enzyme with potent antioxidant properties, as well as the molecular mechanisms involved. 6,7-dihydroxyflavone 60-63 heme oxygenase 1 Homo sapiens 117-121 24503931-2 2014 DHF markedly upregulated HO-1 mRNA and protein expression in human keratinocytes (HaCaT cells), resulting in increased HO-1 activity. 6,7-dihydroxyflavone 0-3 heme oxygenase 1 Homo sapiens 25-29 24503931-2 2014 DHF markedly upregulated HO-1 mRNA and protein expression in human keratinocytes (HaCaT cells), resulting in increased HO-1 activity. 6,7-dihydroxyflavone 0-3 heme oxygenase 1 Homo sapiens 119-123 24503931-3 2014 DHF also increased the protein level of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates HO-1 expression by binding to the antioxidant response element (ARE) within the HO-1 gene promoter, in a time-dependent manner. 6,7-dihydroxyflavone 0-3 heme oxygenase 1 Homo sapiens 129-133 24503931-3 2014 DHF also increased the protein level of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates HO-1 expression by binding to the antioxidant response element (ARE) within the HO-1 gene promoter, in a time-dependent manner. 6,7-dihydroxyflavone 0-3 heme oxygenase 1 Homo sapiens 209-213 24503931-5 2014 DHF activated extracellular-regulated kinase (ERK) and protein kinase B (PKB, Akt) in keratinocytes, while the ERK and Akt inhibitors attenuated DHF-enhanced Nrf2 and HO-1 expression. 6,7-dihydroxyflavone 145-148 heme oxygenase 1 Homo sapiens 167-171 24503931-6 2014 DHF also protected the keratinocytes against hydrogen peroxide- and ultraviolet B-induced oxidative damage, while HO-1, ERK and Akt inhibitors markedly suppressed DHF-mediated cytoprotection. 6,7-dihydroxyflavone 163-166 heme oxygenase 1 Homo sapiens 114-118 25371888-5 2014 qRT-PCR and Western blot analyses showed that diazoxide up-regulated the heme oxygenase-1 (HO-1) expression. Diazoxide 46-55 heme oxygenase 1 Homo sapiens 73-89 25371888-5 2014 qRT-PCR and Western blot analyses showed that diazoxide up-regulated the heme oxygenase-1 (HO-1) expression. Diazoxide 46-55 heme oxygenase 1 Homo sapiens 91-95 25371888-8 2014 CONCLUSION: These results, taken together, strongly suggest that up-regulation of the HO-1 expression is the crucial mechanism responsible for the diazoxide-induced inhibition of the sFlt-1 release and ROS production under hypoxia. Reactive Oxygen Species 202-205 heme oxygenase 1 Homo sapiens 86-90 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. cobaltiprotoporphyrin 30-51 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. cobaltiprotoporphyrin 53-57 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Bilirubin 93-102 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Bilirubin 93-102 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 112-121 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 112-121 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 112-121 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 152-175 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 152-175 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 152-175 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 177-180 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 177-180 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Reactive Oxygen Species 177-180 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 235-257 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 235-257 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 235-257 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 259-266 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 259-266 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. zinc protoporphyrin 259-266 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 294-303 heme oxygenase 1 Homo sapiens 17-21 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 294-303 heme oxygenase 1 Homo sapiens 88-92 25371888-6 2014 In addition, the HO-1 inducer cobalt protoporphyrin (CoPP) and the metabolic product of HO-1 bilirubin mimicked diazoxide to inhibit sFlt-1 release and reactive oxygen species (ROS) production under hypoxia, whereas the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) antagonized the effect of diazoxide. Diazoxide 294-303 heme oxygenase 1 Homo sapiens 88-92 25371888-8 2014 CONCLUSION: These results, taken together, strongly suggest that up-regulation of the HO-1 expression is the crucial mechanism responsible for the diazoxide-induced inhibition of the sFlt-1 release and ROS production under hypoxia. Diazoxide 147-156 heme oxygenase 1 Homo sapiens 86-90 24399465-7 2014 TQ treatments caused increased levels of reactive oxygen species (ROS) and mRNAs of oxidative stress-related genes, NQO1 and HO-1. thymoquinone 0-2 heme oxygenase 1 Homo sapiens 125-129 24633012-1 2014 Haem oxygenase (HO)-1/carbon monoxide (CO) protects cancer cells from oxidative stress, but the gas-responsive signalling mechanisms remain unknown. Cobalt 39-41 heme oxygenase 1 Homo sapiens 0-21 24681888-5 2014 Heme inhibits the transcriptional repressor activity of Bach1, resulting in the derepression of its target genes, such as globin in erythroid cells and heme oxygenase-1 in diverse cell types. Heme 0-4 heme oxygenase 1 Homo sapiens 152-168 24651442-0 2014 Heme oxygenase-1 derived carbon monoxide permits maturation of myeloid cells. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 24690955-2 2014 Bilirubin is produced by heme oxygenase-1 (HO-1), catalyzed by UDP-glucuronosyltransferase (UGT1A1), and has potential as an antioxidant. Bilirubin 0-9 heme oxygenase 1 Homo sapiens 25-41 24690955-2 2014 Bilirubin is produced by heme oxygenase-1 (HO-1), catalyzed by UDP-glucuronosyltransferase (UGT1A1), and has potential as an antioxidant. Bilirubin 0-9 heme oxygenase 1 Homo sapiens 43-47 24355171-9 2014 TQ also generated reactive oxygen species (ROS) and pretreatment with N-acetyl cysteine (NAC) abrogated TQ-induced ROS accumulation, Akt and AMPKalpha activation, Nrf2 nuclear localization, the ARE-luciferase activity, and HO-1 expression in HaCaT cells. Acetylcysteine 70-87 heme oxygenase 1 Homo sapiens 223-227 24681707-5 2014 Two Hsp32-targeting drugs, pegylated zinc protoporphyrine (PEG-ZnPP) and styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP), induced apoptosis and growth arrest in the BCR/ABL1+ cell lines, in Ph- lymphoblastic cell lines and in primary Ph+ and Ph- ALL cells. zinc protoporphyrine 37-57 heme oxygenase 1 Homo sapiens 4-9 24681707-5 2014 Two Hsp32-targeting drugs, pegylated zinc protoporphyrine (PEG-ZnPP) and styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP), induced apoptosis and growth arrest in the BCR/ABL1+ cell lines, in Ph- lymphoblastic cell lines and in primary Ph+ and Ph- ALL cells. peg-znpp 59-67 heme oxygenase 1 Homo sapiens 4-9 24681707-5 2014 Two Hsp32-targeting drugs, pegylated zinc protoporphyrine (PEG-ZnPP) and styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP), induced apoptosis and growth arrest in the BCR/ABL1+ cell lines, in Ph- lymphoblastic cell lines and in primary Ph+ and Ph- ALL cells. 2-Butenedioic acid (2Z)-, polymer with ethenylbenzene 73-92 heme oxygenase 1 Homo sapiens 4-9 24681707-5 2014 Two Hsp32-targeting drugs, pegylated zinc protoporphyrine (PEG-ZnPP) and styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP), induced apoptosis and growth arrest in the BCR/ABL1+ cell lines, in Ph- lymphoblastic cell lines and in primary Ph+ and Ph- ALL cells. zinc protoporphyrin 63-67 heme oxygenase 1 Homo sapiens 4-9 24681707-5 2014 Two Hsp32-targeting drugs, pegylated zinc protoporphyrine (PEG-ZnPP) and styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP), induced apoptosis and growth arrest in the BCR/ABL1+ cell lines, in Ph- lymphoblastic cell lines and in primary Ph+ and Ph- ALL cells. sma-znpp 120-128 heme oxygenase 1 Homo sapiens 4-9 24681707-7 2014 In Ph+ ALL, major growth-inhibitory effects of the Hsp32-targeting drugs were observed in imatinib-sensitive and imatinib-resistant cells. Imatinib Mesylate 90-98 heme oxygenase 1 Homo sapiens 51-56 24681707-7 2014 In Ph+ ALL, major growth-inhibitory effects of the Hsp32-targeting drugs were observed in imatinib-sensitive and imatinib-resistant cells. Imatinib Mesylate 113-121 heme oxygenase 1 Homo sapiens 51-56 24681707-8 2014 Hsp32-targeting drugs were found to synergize with imatinib, nilotinib, and bendamustine in producing growth inhibition and apoptosis in Ph+ ALL cells. Imatinib Mesylate 51-59 heme oxygenase 1 Homo sapiens 0-5 24681707-8 2014 Hsp32-targeting drugs were found to synergize with imatinib, nilotinib, and bendamustine in producing growth inhibition and apoptosis in Ph+ ALL cells. nilotinib 61-70 heme oxygenase 1 Homo sapiens 0-5 24681707-8 2014 Hsp32-targeting drugs were found to synergize with imatinib, nilotinib, and bendamustine in producing growth inhibition and apoptosis in Ph+ ALL cells. Bendamustine Hydrochloride 76-88 heme oxygenase 1 Homo sapiens 0-5 24681707-9 2014 A siRNA against Hsp32 was found to inhibit growth and survival of ALL cells and to synergize with imatinib in suppressing the growth of ALL cells. Imatinib Mesylate 98-106 heme oxygenase 1 Homo sapiens 16-21 24355171-0 2014 Thymoquinone induces heme oxygenase-1 expression in HaCaT cells via Nrf2/ARE activation: Akt and AMPKalpha as upstream targets. thymoquinone 0-12 heme oxygenase 1 Homo sapiens 21-37 24355171-9 2014 TQ also generated reactive oxygen species (ROS) and pretreatment with N-acetyl cysteine (NAC) abrogated TQ-induced ROS accumulation, Akt and AMPKalpha activation, Nrf2 nuclear localization, the ARE-luciferase activity, and HO-1 expression in HaCaT cells. thymoquinone 0-2 heme oxygenase 1 Homo sapiens 223-227 24659985-2 2014 The expression of the heme catabolizing enzyme encoded by this gene, namely HO-1, is required to successfully support reproductive events. Heme 22-26 heme oxygenase 1 Homo sapiens 76-80 24659985-9 2014 In vitro, HO-1 protein levels in AN3 cells augmented after the addition of physiological concentrations of progesterone and estradiol, which confirmed our in vivo observations. Progesterone 107-119 heme oxygenase 1 Homo sapiens 10-14 24659985-9 2014 In vitro, HO-1 protein levels in AN3 cells augmented after the addition of physiological concentrations of progesterone and estradiol, which confirmed our in vivo observations. Estradiol 124-133 heme oxygenase 1 Homo sapiens 10-14 24355171-9 2014 TQ also generated reactive oxygen species (ROS) and pretreatment with N-acetyl cysteine (NAC) abrogated TQ-induced ROS accumulation, Akt and AMPKalpha activation, Nrf2 nuclear localization, the ARE-luciferase activity, and HO-1 expression in HaCaT cells. Acetylcysteine 89-92 heme oxygenase 1 Homo sapiens 223-227 24365205-8 2014 HO-1 increased autophagy, and HO-1 reduced I/R-induced calcium overload in hepatocytes and prevented calpain 2 activation both in vivo and in vitro. Calcium 55-62 heme oxygenase 1 Homo sapiens 30-34 24355171-9 2014 TQ also generated reactive oxygen species (ROS) and pretreatment with N-acetyl cysteine (NAC) abrogated TQ-induced ROS accumulation, Akt and AMPKalpha activation, Nrf2 nuclear localization, the ARE-luciferase activity, and HO-1 expression in HaCaT cells. thymoquinone 104-106 heme oxygenase 1 Homo sapiens 223-227 24355171-10 2014 Taken together, TQ induces HO-1 expression in HaCaT cells by activating Nrf2 through ROS-mediated phosphorylation of Akt and AMPKalpha. Reactive Oxygen Species 85-88 heme oxygenase 1 Homo sapiens 27-31 24407487-4 2014 The (GT)n and (TA)n dinucleotide variations in heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) gene promoters were determined by fragment analysis. Dinucleoside Phosphates 20-32 heme oxygenase 1 Homo sapiens 47-63 24637794-1 2014 Heme oxygenase-1 (HO-1) and its enzymatic by-product carbon monoxide (CO) have emerged as important regulators of acute and chronic inflammation. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 0-16 24637794-1 2014 Heme oxygenase-1 (HO-1) and its enzymatic by-product carbon monoxide (CO) have emerged as important regulators of acute and chronic inflammation. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 18-22 24637794-1 2014 Heme oxygenase-1 (HO-1) and its enzymatic by-product carbon monoxide (CO) have emerged as important regulators of acute and chronic inflammation. Carbon Monoxide 70-72 heme oxygenase 1 Homo sapiens 0-16 24637794-1 2014 Heme oxygenase-1 (HO-1) and its enzymatic by-product carbon monoxide (CO) have emerged as important regulators of acute and chronic inflammation. Carbon Monoxide 70-72 heme oxygenase 1 Homo sapiens 18-22 24407487-4 2014 The (GT)n and (TA)n dinucleotide variations in heme oxygenase 1 (HMOX1) and bilirubin UDP-glucuronosyl transferase (UGT1A1) gene promoters were determined by fragment analysis. Dinucleoside Phosphates 20-32 heme oxygenase 1 Homo sapiens 65-70 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Carbon Monoxide 42-57 heme oxygenase 1 Homo sapiens 76-92 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Carbon Monoxide 42-57 heme oxygenase 1 Homo sapiens 94-98 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Carbon Monoxide 58-60 heme oxygenase 1 Homo sapiens 76-92 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Carbon Monoxide 58-60 heme oxygenase 1 Homo sapiens 94-98 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Pravastatin 252-263 heme oxygenase 1 Homo sapiens 76-92 23972718-4 2014 On the vascular side, the upregulation of carbon monoxide CO, metabolite of heme oxygenase-1 (HO-1; by inhibiting soluble Flt-1, SFlt-1, and soluble endoglin, SEng, release), explains the very promising protective and potentially therapeutic effect of pravastatin in preeclampsia and the paradoxical strong protective effect of cigarette smoking on preeclampsia. Pravastatin 252-263 heme oxygenase 1 Homo sapiens 94-98 24642709-0 2014 Capsaicin ameliorates cisplatin-induced renal injury through induction of heme oxygenase-1. Capsaicin 0-9 heme oxygenase 1 Homo sapiens 74-90 24642709-0 2014 Capsaicin ameliorates cisplatin-induced renal injury through induction of heme oxygenase-1. Cisplatin 22-31 heme oxygenase 1 Homo sapiens 74-90 24642709-4 2014 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in the heme metabolism, has been implicated in a various cellular processes, such as inflammatory injury and anti-oxidant/oxidant homeostasis. Heme 57-61 heme oxygenase 1 Homo sapiens 0-16 24642709-4 2014 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in the heme metabolism, has been implicated in a various cellular processes, such as inflammatory injury and anti-oxidant/oxidant homeostasis. Heme 57-61 heme oxygenase 1 Homo sapiens 18-22 24642709-9 2014 We also found that capsaicin induces HO-1 expression in kidney tissues and HK-2 cells. Capsaicin 19-28 heme oxygenase 1 Homo sapiens 37-41 24642709-10 2014 Notably, the protective effects of capsaicin were completely abrogated by treatment with either the HO inhibitor ZnPP IX or HO-1 knockdown in HK-2 cells. Capsaicin 35-44 heme oxygenase 1 Homo sapiens 124-128 24642709-11 2014 These results suggest that capsaicin has protective effects against cisplatin-induced renal dysfunction through induction of HO-1 as well as inhibition oxidative stress and inflammation. Capsaicin 27-36 heme oxygenase 1 Homo sapiens 125-129 24642709-11 2014 These results suggest that capsaicin has protective effects against cisplatin-induced renal dysfunction through induction of HO-1 as well as inhibition oxidative stress and inflammation. Cisplatin 68-77 heme oxygenase 1 Homo sapiens 125-129 24361488-0 2014 Nrf2/ARE pathway activation, HO-1 and NQO1 induction by polychlorinated biphenyl quinone is associated with reactive oxygen species and PI3K/AKT signaling. polychlorinated biphenyl quinone 56-88 heme oxygenase 1 Homo sapiens 29-33 24503248-2 2014 The Hmox1/carbon monoxide (CO) pathway inhibits soluble Flt-1 (sFlt-1) and soluble Endoglin (sEng). Carbon Monoxide 10-25 heme oxygenase 1 Homo sapiens 4-9 24503248-2 2014 The Hmox1/carbon monoxide (CO) pathway inhibits soluble Flt-1 (sFlt-1) and soluble Endoglin (sEng). Cobalt 27-29 heme oxygenase 1 Homo sapiens 4-9 24623202-10 2014 The TAT-HO-1 attenuated the generation of reactive oxygen species and decreased HaCaT cell radiosensitivity to irradiation. Reactive Oxygen Species 42-65 heme oxygenase 1 Homo sapiens 8-12 24361488-4 2014 In the present study, we found that exposure to PCB29-pQ resulted in a significant increase in Nrf2 and Keap1 expression in total protein, as well as the Nrf2 targeting genes, including NQO1 and HO-1. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 48-56 heme oxygenase 1 Homo sapiens 195-199 24533775-1 2014 BACKGROUND: Previously, we reported that menadione activated rat, native heme oxygenase-2 (HO-2) and human recombinant heme oxygenase-2 selectively; it did not activate spleen, microsomal heme oxygenase-1. Vitamin K 3 41-50 heme oxygenase 1 Homo sapiens 188-204 24361488-8 2014 Additionally, PCB29-pQ treatment led to significant up-regulation of the mRNA level of antioxidant enzymes, NQO1 and HO-1, in a concentration-dependent manner. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 14-22 heme oxygenase 1 Homo sapiens 117-121 24361488-11 2014 Pretreatment with LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K), suppressed the phosphorylation of AKT and inhibited PCB29-pQ induced Nrf2/HO-1 activation, meanwhile, GSK-3beta expression was increased accordingly. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 18-26 heme oxygenase 1 Homo sapiens 164-168 24361488-11 2014 Pretreatment with LY294002, a specific inhibitor of phosphatidylinositol 3-kinase (PI3K), suppressed the phosphorylation of AKT and inhibited PCB29-pQ induced Nrf2/HO-1 activation, meanwhile, GSK-3beta expression was increased accordingly. 2,3,5-trichloro-6-phenyl-(1,4)benzoquinone 142-150 heme oxygenase 1 Homo sapiens 164-168 24361900-0 2014 Nitric oxide sets off an antioxidant response in adrenal cells: involvement of sGC and Nrf2 in HO-1 induction. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 95-99 24361900-2 2014 In adrenal cells, HO-1 induction by ACTH exerts a modulatory effect on steroid production as well. Steroids 71-78 heme oxygenase 1 Homo sapiens 18-22 24361900-3 2014 As nitric oxide (NO) has been also regarded as an autocrine/paracrine modulator of adrenal steroidogenesis we sought to study the effects of NO on the induction of HO-1 and the mechanism involved. Nitric Oxide 3-15 heme oxygenase 1 Homo sapiens 164-168 24361900-4 2014 We hereby analyzed the time and dose-dependent effect of a NO-donor (DETA/NO) on HO-1 induction in a murine adrenocortical cell line. DEET 69-73 heme oxygenase 1 Homo sapiens 81-85 24361900-8 2014 We finally show that the effects of the NO-donor are reproduced by a permeable analog of cGMP and that a soluble guanylate cyclase specific inhibitor blocked both the induction of HO-1 by NO and the nuclear translocation of Nrf2. Cyclic GMP 89-93 heme oxygenase 1 Homo sapiens 180-184 24516582-7 2014 Interestingly, the most affected biological pathway by both 2 or 12 hrs of iAs exposure were the Nrf2-Keap1 pathway, represented by the highly up-regulated HMOX1 transcript, which is transcriptionally regulated by the transcription factor Nrf2. 4-Iodoacetamidosalicylic acid 75-78 heme oxygenase 1 Homo sapiens 156-161 24516582-10 2014 Since HMOX1 expression increased early (20 min) and was responsive to low iAs concentrations (0.1 microM), this gene could be a suitable early biomarker for iAs exposure. 4-Iodoacetamidosalicylic acid 74-77 heme oxygenase 1 Homo sapiens 6-11 24516582-10 2014 Since HMOX1 expression increased early (20 min) and was responsive to low iAs concentrations (0.1 microM), this gene could be a suitable early biomarker for iAs exposure. 4-Iodoacetamidosalicylic acid 157-160 heme oxygenase 1 Homo sapiens 6-11 23852531-0 2014 Hydrogen sulfide induces heme oxygenase-1 in human kidney cells. Hydrogen Sulfide 0-16 heme oxygenase 1 Homo sapiens 25-41 23852531-2 2014 The aim of this study was to investigate if H2S effects the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in human kidney cells. Hydrogen Sulfide 44-47 heme oxygenase 1 Homo sapiens 97-113 23852531-2 2014 The aim of this study was to investigate if H2S effects the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in human kidney cells. Hydrogen Sulfide 44-47 heme oxygenase 1 Homo sapiens 115-119 23852531-5 2014 There was a significant increase in HO-1 expression after treatment with the H2S donors in both mesangial and podocyte cells. Hydrogen Sulfide 77-80 heme oxygenase 1 Homo sapiens 36-40 23852531-6 2014 These results suggest that H2S has a role in the regulation of HO-1 expression, and the ability to upregulate this antioxidant enzyme maybe a potential mechanism by which H2S exerts its protective effects. Hydrogen Sulfide 27-30 heme oxygenase 1 Homo sapiens 63-67 23852531-6 2014 These results suggest that H2S has a role in the regulation of HO-1 expression, and the ability to upregulate this antioxidant enzyme maybe a potential mechanism by which H2S exerts its protective effects. Hydrogen Sulfide 171-174 heme oxygenase 1 Homo sapiens 63-67 24396429-5 2014 Correlation analysis showed that the expression levels of HO-1 protein were positively correlated with the HO-1 mRNA expression levels (r=0.680; P=0.000), and the levels of ROS (r=0.572; P=0.000) and MDA (r=0.614; P=0.000). Reactive Oxygen Species 173-176 heme oxygenase 1 Homo sapiens 58-62 24396429-6 2014 HO-1 mRNA expression levels were found to positively correlate with the levels of MDA (r=0.451; P=0.010) and fasting plasma glucose (FPG; r=0.337; P=0.039). Glucose 124-131 heme oxygenase 1 Homo sapiens 0-4 24396429-7 2014 Partial correlation analysis demonstrated that, after removing the effects of body mass index, FPG and 2-h plasma glucose, HO-1 protein expression levels were positively correlated with the levels of HO-1 mRNA expression (r=0.611; P=0.005), ROS (r=0.526; P=0.021) and MDA (r=0.519; P=0.015). Reactive Oxygen Species 241-244 heme oxygenase 1 Homo sapiens 123-127 24396429-7 2014 Partial correlation analysis demonstrated that, after removing the effects of body mass index, FPG and 2-h plasma glucose, HO-1 protein expression levels were positively correlated with the levels of HO-1 mRNA expression (r=0.611; P=0.005), ROS (r=0.526; P=0.021) and MDA (r=0.519; P=0.015). Reactive Oxygen Species 241-244 heme oxygenase 1 Homo sapiens 200-204 24396429-8 2014 These findings indicate that pregnant females with GDM may be protected against oxidative injury due to the induction of adaptive and compensatory expression of HO-1 to guard against oxidative stress induced by high glucose levels. Glucose 216-223 heme oxygenase 1 Homo sapiens 161-165 24451142-0 2014 Resveratrol partially prevents rotenone-induced neurotoxicity in dopaminergic SH-SY5Y cells through induction of heme oxygenase-1 dependent autophagy. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 113-129 23934646-0 2014 Neuroprotection by monocarbonyl dimethoxycurcumin C: ameliorating the toxicity of mutant TDP-43 via HO-1. monocarbonyl dimethoxycurcumin c 19-51 heme oxygenase 1 Homo sapiens 100-104 23934646-9 2014 Znpp, which is known an inhibitor of HO-1, dramatically interfered with the function of Compound C. In addition, Compound C was tested in vivo, and HO-1 was significantly upregulated at the hippocampus. zinc protoporphyrin 0-4 heme oxygenase 1 Homo sapiens 37-41 23934646-9 2014 Znpp, which is known an inhibitor of HO-1, dramatically interfered with the function of Compound C. In addition, Compound C was tested in vivo, and HO-1 was significantly upregulated at the hippocampus. zinc protoporphyrin 0-4 heme oxygenase 1 Homo sapiens 148-152 24489685-3 2014 Among the three organosulfur compounds, DATS was found to be most potent in inducing heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO1) in human gastric epithelial (AGS) cells. dats 40-44 heme oxygenase 1 Homo sapiens 85-101 24302760-7 2014 Tan treatment induced the phosphorylation and nuclear translocation of Nrf2 and subsequently increased the expression of HO-1, and these effects were abolished by the specific ERK inhibitors, PD98059 and U0126. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 192-199 heme oxygenase 1 Homo sapiens 121-125 24302760-7 2014 Tan treatment induced the phosphorylation and nuclear translocation of Nrf2 and subsequently increased the expression of HO-1, and these effects were abolished by the specific ERK inhibitors, PD98059 and U0126. U 0126 204-209 heme oxygenase 1 Homo sapiens 121-125 24302760-8 2014 Moreover, HO-1 small interfering RNA or zinc protoporphyrin (a HO-1 inhibitor) abrogated Tan-mediated suppression of lipid accumulation in macrophages. zinc protoporphyrin 40-59 heme oxygenase 1 Homo sapiens 63-67 24302760-9 2014 Our current findings demonstrate that a novel HO-1-dependent mechanism is involved in the regulation of cholesterol balance by Tan. Cholesterol 104-115 heme oxygenase 1 Homo sapiens 46-50 24355260-2 2014 Of interest, patients with lung tumors have been noted to have an increase in endogenous carbon monoxide production via upregulation of hemeoxygenase-1 activity. Carbon Monoxide 89-104 heme oxygenase 1 Homo sapiens 136-151 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. Heme 59-63 heme oxygenase 1 Homo sapiens 0-4 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. Biliverdine 127-140 heme oxygenase 1 Homo sapiens 0-4 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. bilirubin-ix 152-164 heme oxygenase 1 Homo sapiens 0-4 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. Carbon Monoxide 226-241 heme oxygenase 1 Homo sapiens 0-4 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. Carbon Monoxide 243-245 heme oxygenase 1 Homo sapiens 0-4 24095978-4 2014 HO-1/BVR-A reduces the intracellular levels of pro-oxidant heme and generates equimolar amounts of the free radical scavengers biliverdin-IX alpha (BV)/bilirubin-IX alpha (BR) as well as the pleiotropic gaseous neuromodulator carbon monoxide (CO) and ferrous iron. ferrous iron 251-263 heme oxygenase 1 Homo sapiens 0-4 24434148-10 2014 NADPH oxidase inhibitor Apocynin and superoxide scavenger N-acetyl-cysteine (NAC) significantly attenuated EI-stimulated expression of OKL38 and HO-1. acetovanillone 24-32 heme oxygenase 1 Homo sapiens 145-149 24434148-10 2014 NADPH oxidase inhibitor Apocynin and superoxide scavenger N-acetyl-cysteine (NAC) significantly attenuated EI-stimulated expression of OKL38 and HO-1. Superoxides 37-47 heme oxygenase 1 Homo sapiens 145-149 24434148-10 2014 NADPH oxidase inhibitor Apocynin and superoxide scavenger N-acetyl-cysteine (NAC) significantly attenuated EI-stimulated expression of OKL38 and HO-1. Acetylcysteine 58-75 heme oxygenase 1 Homo sapiens 145-149 24434148-10 2014 NADPH oxidase inhibitor Apocynin and superoxide scavenger N-acetyl-cysteine (NAC) significantly attenuated EI-stimulated expression of OKL38 and HO-1. Acetylcysteine 77-80 heme oxygenase 1 Homo sapiens 145-149 24434148-10 2014 NADPH oxidase inhibitor Apocynin and superoxide scavenger N-acetyl-cysteine (NAC) significantly attenuated EI-stimulated expression of OKL38 and HO-1. aziridine 107-109 heme oxygenase 1 Homo sapiens 145-149 24300974-0 2014 Gambogic acid synergistically potentiates cisplatin-induced apoptosis in non-small-cell lung cancer through suppressing NF-kappaB and MAPK/HO-1 signalling. gambogic acid 0-13 heme oxygenase 1 Homo sapiens 139-143 24451142-0 2014 Resveratrol partially prevents rotenone-induced neurotoxicity in dopaminergic SH-SY5Y cells through induction of heme oxygenase-1 dependent autophagy. Rotenone 31-39 heme oxygenase 1 Homo sapiens 113-129 24300974-0 2014 Gambogic acid synergistically potentiates cisplatin-induced apoptosis in non-small-cell lung cancer through suppressing NF-kappaB and MAPK/HO-1 signalling. Cisplatin 42-51 heme oxygenase 1 Homo sapiens 139-143 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. Cisplatin 44-48 heme oxygenase 1 Homo sapiens 131-147 24451142-9 2014 Further, rotenone reduced heme oxygenase-1 (HO-1) expression, whereas resveratrol increased HO-1 expression. Rotenone 9-17 heme oxygenase 1 Homo sapiens 26-42 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. Cisplatin 44-48 heme oxygenase 1 Homo sapiens 149-153 24451142-9 2014 Further, rotenone reduced heme oxygenase-1 (HO-1) expression, whereas resveratrol increased HO-1 expression. Rotenone 9-17 heme oxygenase 1 Homo sapiens 44-48 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. gambogic acid 60-62 heme oxygenase 1 Homo sapiens 131-147 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. gambogic acid 60-62 heme oxygenase 1 Homo sapiens 149-153 24451142-9 2014 Further, rotenone reduced heme oxygenase-1 (HO-1) expression, whereas resveratrol increased HO-1 expression. Resveratrol 70-81 heme oxygenase 1 Homo sapiens 92-96 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. Reactive Oxygen Species 212-215 heme oxygenase 1 Homo sapiens 149-153 24300974-8 2014 Importantly, it was found that, followed by CDDP treatment, GA could inhibit NF-kappaB and mitogen-activated protein kinase (MAPK)/heme oxygenase-1 (HO-1) signalling pathways, which have been validated to reduce ROS release and confer CDDP resistance. Cisplatin 235-239 heme oxygenase 1 Homo sapiens 149-153 24300974-10 2014 Moreover, our results indicated that the combination of CDDP and GA exerted increased antitumour effects on A549 xenograft models through inhibiting NF-kappaB, HO-1, and subsequently inducing apoptosis. Cisplatin 56-60 heme oxygenase 1 Homo sapiens 160-164 24300974-10 2014 Moreover, our results indicated that the combination of CDDP and GA exerted increased antitumour effects on A549 xenograft models through inhibiting NF-kappaB, HO-1, and subsequently inducing apoptosis. gambogic acid 65-67 heme oxygenase 1 Homo sapiens 160-164 24300974-11 2014 CONCLUSION: Gambogic acid sensitises lung cancer cells to CDDP in vitro and in vivo in NSCLC through inactivation of NF-kappaB and MAPK/HO-1 signalling pathways, providing a rationale for the combined use of CDDP and GA in lung cancer chemotherapy. gambogic acid 12-25 heme oxygenase 1 Homo sapiens 136-140 24451142-10 2014 Pharmacological inhibition of HO-1 abolished resveratrol-mediated autophagy and neuroprotection. Resveratrol 45-56 heme oxygenase 1 Homo sapiens 30-34 24451142-11 2014 Notably, the effects of a pharmacological inducer of HO-1 were similar to those of resveratrol, and protected against rotenone-induced cell death in an autophagy-dependent manner, validating the hypothesis of HO-1 dependent autophagy in preventing neuronal death in the in vitro PD model. Rotenone 118-126 heme oxygenase 1 Homo sapiens 53-57 24451142-11 2014 Notably, the effects of a pharmacological inducer of HO-1 were similar to those of resveratrol, and protected against rotenone-induced cell death in an autophagy-dependent manner, validating the hypothesis of HO-1 dependent autophagy in preventing neuronal death in the in vitro PD model. Rotenone 118-126 heme oxygenase 1 Homo sapiens 209-213 24451142-12 2014 Collectively, our findings suggest that resveratrol induces HO-1 expression and prevents dopaminergic cell death by regulating autophagic flux; thus protecting against rotenone-induced neuronal apoptosis. Resveratrol 40-51 heme oxygenase 1 Homo sapiens 60-64 24183966-8 2014 HO-1 may also contribute to the iron accumulation in neurons, but its mechanism needs to be further investigated. Iron 32-36 heme oxygenase 1 Homo sapiens 0-4 24035224-6 2014 In addition, sauchinone induced heme oxygenase (HO)-1 expression through nuclear translocation of nuclear factor E2-related factor 2 in HUVEC. sauchinone 13-23 heme oxygenase 1 Homo sapiens 32-53 24239896-4 2014 The induction of HO-1 was associated with an increase in NF-E2-related factor-2 (Nrf2) activity and reactive oxygen species (ROS). Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 17-21 24239896-4 2014 The induction of HO-1 was associated with an increase in NF-E2-related factor-2 (Nrf2) activity and reactive oxygen species (ROS). Reactive Oxygen Species 125-128 heme oxygenase 1 Homo sapiens 17-21 24239896-6 2014 In addition, the CM-mediated induction of HO-1 and activation of Nrf2 was abolished by acetylcysteine. Acetylcysteine 87-101 heme oxygenase 1 Homo sapiens 42-46 24239896-9 2014 Thus, induction of HO-1 via the ROS-Nrf2 pathway counteracts the anti-proliferative and inflammatory actions of CM. Reactive Oxygen Species 32-35 heme oxygenase 1 Homo sapiens 19-23 24239896-3 2014 We found that three distinct CM, including high-osmolar (diatrizoate), low-osmolar (iopamidol), and iso-osmolar (iodixanol), stimulated the expression of HO-1 protein and mRNA. Diatrizoate 57-68 heme oxygenase 1 Homo sapiens 154-158 24239896-3 2014 We found that three distinct CM, including high-osmolar (diatrizoate), low-osmolar (iopamidol), and iso-osmolar (iodixanol), stimulated the expression of HO-1 protein and mRNA. Iopamidol 84-93 heme oxygenase 1 Homo sapiens 154-158 24239896-3 2014 We found that three distinct CM, including high-osmolar (diatrizoate), low-osmolar (iopamidol), and iso-osmolar (iodixanol), stimulated the expression of HO-1 protein and mRNA. iodixanol 113-122 heme oxygenase 1 Homo sapiens 154-158 24089372-3 2014 Heme arginate (HA), a HO-1 inducer, is a promising, translatable, preconditioning agent. heme arginate 0-13 heme oxygenase 1 Homo sapiens 22-26 24126012-0 2014 Mechanistic study on the biological effects of silver and gold nanoparticles in Caco-2 cells--induction of the Nrf2/HO-1 pathway by high concentrations of silver nanoparticles. Silver 47-53 heme oxygenase 1 Homo sapiens 116-120 24126012-0 2014 Mechanistic study on the biological effects of silver and gold nanoparticles in Caco-2 cells--induction of the Nrf2/HO-1 pathway by high concentrations of silver nanoparticles. Silver 155-161 heme oxygenase 1 Homo sapiens 116-120 24126012-5 2014 Furthermore, siRNA silencing of Nrf2 transcripts significantly reduced the AgNP-induced HO-1 mRNA induction, suggesting a key role for Nrf2 in the control of HO-1 expression. agnp 75-79 heme oxygenase 1 Homo sapiens 88-92 24126012-5 2014 Furthermore, siRNA silencing of Nrf2 transcripts significantly reduced the AgNP-induced HO-1 mRNA induction, suggesting a key role for Nrf2 in the control of HO-1 expression. agnp 75-79 heme oxygenase 1 Homo sapiens 158-162 24089372-3 2014 Heme arginate (HA), a HO-1 inducer, is a promising, translatable, preconditioning agent. heme arginate 15-17 heme oxygenase 1 Homo sapiens 22-26 23861314-6 2014 Interestingly, protoporphyrin-IX, a competitive inhibitor of HO-1 activity, partly attenuated the inhibitory effects of Res and Pic (but not TMS) on TNF-alpha and IL-1beta production, suggesting that suppression of TNF-alpha and IL-1beta production correlates at least in part with HO-1 expression. protoporphyrin IX 15-32 heme oxygenase 1 Homo sapiens 61-65 25344086-4 2014 HO-1 along with its reaction products bilirubin and CO are protective against ischemia-induced injury (myocardial infarction, ischemia-reperfusion (IR)-injury and post-infarct structural remodelling). Bilirubin 38-47 heme oxygenase 1 Homo sapiens 0-4 25344086-4 2014 HO-1 along with its reaction products bilirubin and CO are protective against ischemia-induced injury (myocardial infarction, ischemia-reperfusion (IR)-injury and post-infarct structural remodelling). Carbon Monoxide 52-54 heme oxygenase 1 Homo sapiens 0-4 23861314-6 2014 Interestingly, protoporphyrin-IX, a competitive inhibitor of HO-1 activity, partly attenuated the inhibitory effects of Res and Pic (but not TMS) on TNF-alpha and IL-1beta production, suggesting that suppression of TNF-alpha and IL-1beta production correlates at least in part with HO-1 expression. protoporphyrin IX 15-32 heme oxygenase 1 Homo sapiens 282-286 24592391-0 2014 Human pharmacokinetics of high dose oral curcumin and its effect on heme oxygenase-1 expression in healthy male subjects. Curcumin 41-49 heme oxygenase 1 Homo sapiens 68-84 24060752-5 2014 RESULTS: The sulforaphane treatment activated Nrf2, increased levels of the Nrf2 target heme oxygenase-1 and subsequently lowered oxidant stress as shown by the decline in lipid peroxidation and 3-nitrotyrosine protein adducts and an increase in GSH levels after the acute ethanol treatment. sulforaphane 13-25 heme oxygenase 1 Homo sapiens 88-104 25273386-8 2014 Tryptanthrin also up-regulated the expression of the heme oxygenase 1 and glutamate-cysteine ligase catalytic subunits, which are representative target genes of Nrf2. tryptanthrine 0-12 heme oxygenase 1 Homo sapiens 53-69 24592391-3 2014 We investigated the inducibility of HO-1 by orally administered curcumin in healthy male subjects and its correlation with the GT length polymorphism. Curcumin 64-72 heme oxygenase 1 Homo sapiens 36-40 24117426-0 2014 Andrographolide exerts anti-hepatitis C virus activity by up-regulating haeme oxygenase-1 via the p38 MAPK/Nrf2 pathway in human hepatoma cells. andrographolide 0-15 heme oxygenase 1 Homo sapiens 72-89 24117426-7 2014 Andrographolide up-regulated the expression of haeme oxygenase-1 (HO-1), leading to increased amounts of its metabolite biliverdin, which was found to suppress HCV replication by promoting the antiviral IFN responses and inhibiting NS3/4A protease activity. andrographolide 0-15 heme oxygenase 1 Homo sapiens 47-64 24117426-9 2014 Andrographolide activated p38 MAPK phosphorylation, which stimulated nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated HO-1 expression, and this was found to be associated with its anti-HCV activity. andrographolide 0-15 heme oxygenase 1 Homo sapiens 129-133 24117426-10 2014 CONCLUSIONS AND IMPLICATIONS: Our results demonstrate that andrographolide has the potential to control HCV replication and suggest that targeting the Nrf2-HO-1 signalling pathway might be a promising strategy for drug development. andrographolide 59-74 heme oxygenase 1 Homo sapiens 156-160 24117426-7 2014 Andrographolide up-regulated the expression of haeme oxygenase-1 (HO-1), leading to increased amounts of its metabolite biliverdin, which was found to suppress HCV replication by promoting the antiviral IFN responses and inhibiting NS3/4A protease activity. andrographolide 0-15 heme oxygenase 1 Homo sapiens 66-70 24117426-7 2014 Andrographolide up-regulated the expression of haeme oxygenase-1 (HO-1), leading to increased amounts of its metabolite biliverdin, which was found to suppress HCV replication by promoting the antiviral IFN responses and inhibiting NS3/4A protease activity. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 47-64 24117426-7 2014 Andrographolide up-regulated the expression of haeme oxygenase-1 (HO-1), leading to increased amounts of its metabolite biliverdin, which was found to suppress HCV replication by promoting the antiviral IFN responses and inhibiting NS3/4A protease activity. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 66-70 24117426-8 2014 Significantly, these antiviral effects were attenuated by an HO-1-specific inhibitor or HO-1 gene knockdown, indicating that HO-1 contributed to the anti-HCV activity of andrographolide. andrographolide 170-185 heme oxygenase 1 Homo sapiens 61-65 24117426-8 2014 Significantly, these antiviral effects were attenuated by an HO-1-specific inhibitor or HO-1 gene knockdown, indicating that HO-1 contributed to the anti-HCV activity of andrographolide. andrographolide 170-185 heme oxygenase 1 Homo sapiens 88-92 24117426-8 2014 Significantly, these antiviral effects were attenuated by an HO-1-specific inhibitor or HO-1 gene knockdown, indicating that HO-1 contributed to the anti-HCV activity of andrographolide. andrographolide 170-185 heme oxygenase 1 Homo sapiens 88-92 24766133-3 2014 During the last decade, different strategies such as pharmacologic or gene therapy modulation of heme oxygenase-1 (HO-1) and the administration of its metabolic product, carbon monoxide (CO), have been shown to display beneficial immunoregulatory and cytoprotective properties. Carbon Monoxide 187-189 heme oxygenase 1 Homo sapiens 97-113 25146616-14 2014 According to progression of CML, HO-1 expression level in CML patients increased from CP (0.009 5+-0.017 6) to AP (0.028 0+-0.055 7) and then to BP (0.276 7 +- 0.447 0). Benzo(a)pyrene 145-147 heme oxygenase 1 Homo sapiens 33-37 24655000-7 2014 The water-soluble forms of the HO-1 inhibitor Zinc protoporphyrin (ZnPP) have seemed promising in different in-vivo models, in which it has induced growth arrest in tumour cells with few, if any, side effects. Water 4-9 heme oxygenase 1 Homo sapiens 31-35 24655000-7 2014 The water-soluble forms of the HO-1 inhibitor Zinc protoporphyrin (ZnPP) have seemed promising in different in-vivo models, in which it has induced growth arrest in tumour cells with few, if any, side effects. zinc protoporphyrin 46-65 heme oxygenase 1 Homo sapiens 31-35 24655000-7 2014 The water-soluble forms of the HO-1 inhibitor Zinc protoporphyrin (ZnPP) have seemed promising in different in-vivo models, in which it has induced growth arrest in tumour cells with few, if any, side effects. zinc protoporphyrin 67-71 heme oxygenase 1 Homo sapiens 31-35 24934350-0 2014 Salvianolic acid B inhibits atherogenesis of vascular cells through induction of Nrf2-dependent heme oxygenase-1. salvianolic acid B 0-18 heme oxygenase 1 Homo sapiens 96-112 24934350-4 2014 METHODS AND RESULTS: Treatment of vascular smooth muscle cells with Sal B effectively inhibited platelet-derived growth factor (PDGF)-induced cell proliferation and migration, and markedly increased heme oxygenase-1 (HO-1) expression. salvianolic acid B 68-73 heme oxygenase 1 Homo sapiens 199-215 24934350-4 2014 METHODS AND RESULTS: Treatment of vascular smooth muscle cells with Sal B effectively inhibited platelet-derived growth factor (PDGF)-induced cell proliferation and migration, and markedly increased heme oxygenase-1 (HO-1) expression. salvianolic acid B 68-73 heme oxygenase 1 Homo sapiens 217-221 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Heme 70-74 heme oxygenase 1 Homo sapiens 88-92 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Biliverdine 165-175 heme oxygenase 1 Homo sapiens 70-86 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Biliverdine 165-175 heme oxygenase 1 Homo sapiens 88-92 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Iron 187-191 heme oxygenase 1 Homo sapiens 70-86 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Iron 187-191 heme oxygenase 1 Homo sapiens 88-92 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Carbon Monoxide 207-222 heme oxygenase 1 Homo sapiens 70-86 24934350-7 2014 Knockdown of HO-1 expression by siRNA abolished the effects of Sal B in vascular cells and prevented the inhibition of proliferation, migration, and inflammation in HO-1-deficient cells. salvianolic acid B 63-68 heme oxygenase 1 Homo sapiens 13-17 24934350-9 2014 CONCLUSIONS: We conclude that Sal B exerts antiatherogenic effects by inhibiting the proliferation, migration, and inflammation of vascular cells through induction of HO-1 via Nrf2 activation. salvianolic acid B 30-35 heme oxygenase 1 Homo sapiens 167-171 24275768-1 2014 PURPOSE OF REVIEW: Heme oxygenase activity, possessed by an inducible heme oxygenase-1 (HO-1) and a constitutive isoform (HO-2), catalyzes the conversion of heme to biliverdin, liberates iron, and generates carbon monoxide. Carbon Monoxide 207-222 heme oxygenase 1 Homo sapiens 88-92 24766133-3 2014 During the last decade, different strategies such as pharmacologic or gene therapy modulation of heme oxygenase-1 (HO-1) and the administration of its metabolic product, carbon monoxide (CO), have been shown to display beneficial immunoregulatory and cytoprotective properties. Carbon Monoxide 187-189 heme oxygenase 1 Homo sapiens 115-119 25530680-7 2014 We report that Mox-LDL inhibits endothelial cell motility and tubulogenesis through an increase in miR-22 and heme oxygenase 1 expression. mox-ldl 15-22 heme oxygenase 1 Homo sapiens 110-126 25762501-7 2014 We also show that ER stress combined with inflammation synergistically upregulated the expression of the iron carrier protein NGAL and the stress-inducible heme degrading enzyme heme oxygenase-1 (HO-1) leading to iron liberation. Heme 156-160 heme oxygenase 1 Homo sapiens 178-194 25762501-7 2014 We also show that ER stress combined with inflammation synergistically upregulated the expression of the iron carrier protein NGAL and the stress-inducible heme degrading enzyme heme oxygenase-1 (HO-1) leading to iron liberation. Iron 213-217 heme oxygenase 1 Homo sapiens 178-194 24763872-0 2014 Cytoprotective effect of beta-lapachone by inducing heme oxygenase-1 expression and AMP-activated protein kinase activation in human endothelial cells. beta-lapachone 25-39 heme oxygenase 1 Homo sapiens 52-68 24763872-11 2014 Cytoprotection by BL was almost completely abolished by CC and DC and partly by SnPP, a competitive inhibitor of HO-1. S-Nitroso-N-propionyl-D,L-penicillamine 80-84 heme oxygenase 1 Homo sapiens 113-117 25386047-0 2014 Epigallocatechin gallate attenuates proliferation and oxidative stress in human vascular smooth muscle cells induced by interleukin-1beta via heme oxygenase-1. epigallocatechin gallate 0-24 heme oxygenase 1 Homo sapiens 142-158 25386047-3 2014 This study aimed to elucidate the effects of the most active green tea catechin derivative, (-)-epigallocatechin-3-gallate (EGCG), in human aortic smooth muscle cells (HASMCs), focusing particularly on the role of a potent anti-inflammatory and antioxidative enzyme heme oxygenase-1 (HO-1). epigallocatechin gallate 92-122 heme oxygenase 1 Homo sapiens 266-282 25386047-3 2014 This study aimed to elucidate the effects of the most active green tea catechin derivative, (-)-epigallocatechin-3-gallate (EGCG), in human aortic smooth muscle cells (HASMCs), focusing particularly on the role of a potent anti-inflammatory and antioxidative enzyme heme oxygenase-1 (HO-1). epigallocatechin gallate 92-122 heme oxygenase 1 Homo sapiens 284-288 25386047-3 2014 This study aimed to elucidate the effects of the most active green tea catechin derivative, (-)-epigallocatechin-3-gallate (EGCG), in human aortic smooth muscle cells (HASMCs), focusing particularly on the role of a potent anti-inflammatory and antioxidative enzyme heme oxygenase-1 (HO-1). epigallocatechin gallate 124-128 heme oxygenase 1 Homo sapiens 266-282 25386047-3 2014 This study aimed to elucidate the effects of the most active green tea catechin derivative, (-)-epigallocatechin-3-gallate (EGCG), in human aortic smooth muscle cells (HASMCs), focusing particularly on the role of a potent anti-inflammatory and antioxidative enzyme heme oxygenase-1 (HO-1). epigallocatechin gallate 124-128 heme oxygenase 1 Homo sapiens 284-288 25386047-4 2014 We found that pretreatment of EGCG dose- and time-dependently induced HO-1 protein levels in HASMCs. epigallocatechin gallate 30-34 heme oxygenase 1 Homo sapiens 70-74 25386047-6 2014 The HO-1 inducer CoPPIX decreased IL-1beta-induced cell proliferation, whereas the HO-1 enzyme inhibitor ZnPPIX significantly reversed EGCG-caused growth inhibition in IL-1beta-treated HASMCs. coppix 17-23 heme oxygenase 1 Homo sapiens 4-8 25386047-6 2014 The HO-1 inducer CoPPIX decreased IL-1beta-induced cell proliferation, whereas the HO-1 enzyme inhibitor ZnPPIX significantly reversed EGCG-caused growth inhibition in IL-1beta-treated HASMCs. zinc protoporphyrin 105-111 heme oxygenase 1 Homo sapiens 83-87 25386047-6 2014 The HO-1 inducer CoPPIX decreased IL-1beta-induced cell proliferation, whereas the HO-1 enzyme inhibitor ZnPPIX significantly reversed EGCG-caused growth inhibition in IL-1beta-treated HASMCs. epigallocatechin gallate 135-139 heme oxygenase 1 Homo sapiens 83-87 25386047-6 2014 The HO-1 inducer CoPPIX decreased IL-1beta-induced cell proliferation, whereas the HO-1 enzyme inhibitor ZnPPIX significantly reversed EGCG-caused growth inhibition in IL-1beta-treated HASMCs. hasmcs 185-191 heme oxygenase 1 Homo sapiens 83-87 25386047-8 2014 These results suggest that EGCG might serve as a complementary and alternative medicine in the treatment of these pathologies by inducing HO-1 expression and subsequently decreasing VSMC proliferation. epigallocatechin gallate 27-31 heme oxygenase 1 Homo sapiens 138-142 24220687-10 2014 We also demonstrated that sanguinarine induced HO-1 expression, and that the inhibition of MMP-9 and COX-2 expression and the enzymatic activity of sanguinarine were abrogated by siRNA-mediated knockdown of HO-1 expression. sanguinarine 26-38 heme oxygenase 1 Homo sapiens 47-51 24447094-0 2014 PPARalpha activation sensitizes cancer cells to epigallocatechin-3-gallate (EGCG) treatment via suppressing heme oxygenase-1. epigallocatechin gallate 48-74 heme oxygenase 1 Homo sapiens 108-124 24447094-0 2014 PPARalpha activation sensitizes cancer cells to epigallocatechin-3-gallate (EGCG) treatment via suppressing heme oxygenase-1. epigallocatechin gallate 76-80 heme oxygenase 1 Homo sapiens 108-124 24447094-6 2014 The PPARalpha agonist clofibrate blocked cytoprotective heme oxygenase-1 (HO-1) induction and sensitized multiple types of cancer cells to EGCG-induced cell death. Clofibrate 22-32 heme oxygenase 1 Homo sapiens 56-72 24447094-6 2014 The PPARalpha agonist clofibrate blocked cytoprotective heme oxygenase-1 (HO-1) induction and sensitized multiple types of cancer cells to EGCG-induced cell death. Clofibrate 22-32 heme oxygenase 1 Homo sapiens 74-78 24447094-7 2014 Conversely, the PPARalpha inhibitor G6471 and PPARalpha siRNA increased HO-1 expression. g6471 36-41 heme oxygenase 1 Homo sapiens 72-76 24447094-9 2014 Moreover, cell death induced by EGCG plus clofibrate was partially reversed by HO-1 overexpression in PANC1 cells. epigallocatechin gallate 32-36 heme oxygenase 1 Homo sapiens 79-83 24447094-9 2014 Moreover, cell death induced by EGCG plus clofibrate was partially reversed by HO-1 overexpression in PANC1 cells. Clofibrate 42-52 heme oxygenase 1 Homo sapiens 79-83 24447094-10 2014 These results indicate that PPARalpha is a direct and negative regulator of HO-1 induced by EGCG and confers cell susceptibility to EGCG. epigallocatechin gallate 92-96 heme oxygenase 1 Homo sapiens 76-80 24447094-10 2014 These results indicate that PPARalpha is a direct and negative regulator of HO-1 induced by EGCG and confers cell susceptibility to EGCG. epigallocatechin gallate 132-136 heme oxygenase 1 Homo sapiens 76-80 25150313-0 2014 MiR-200c sensitizes clear-cell renal cell carcinoma cells to sorafenib and imatinib by targeting heme oxygenase-1. Imatinib Mesylate 75-83 heme oxygenase 1 Homo sapiens 97-113 25150313-5 2014 We provided evidences to elucidate that miR-200c could sensitize ccRCC cells to sorafenib or imatinib to inhibit cell proliferation, at least partly by targeting HO-1. Sorafenib 80-89 heme oxygenase 1 Homo sapiens 162-166 25150313-5 2014 We provided evidences to elucidate that miR-200c could sensitize ccRCC cells to sorafenib or imatinib to inhibit cell proliferation, at least partly by targeting HO-1. Imatinib Mesylate 93-101 heme oxygenase 1 Homo sapiens 162-166 24220687-0 2014 Sanguinarine inhibits invasiveness and the MMP-9 and COX-2 expression in TPA-induced breast cancer cells by inducing HO-1 expression. sanguinarine 0-12 heme oxygenase 1 Homo sapiens 117-121 24220687-0 2014 Sanguinarine inhibits invasiveness and the MMP-9 and COX-2 expression in TPA-induced breast cancer cells by inducing HO-1 expression. Tetradecanoylphorbol Acetate 73-76 heme oxygenase 1 Homo sapiens 117-121 24220687-4 2014 In the present study, we investigated the anti-invasive mechanism of sanguinarine by focusing on its role in inducing HO-1 in breast cancer cells. sanguinarine 69-81 heme oxygenase 1 Homo sapiens 118-122 24220687-10 2014 We also demonstrated that sanguinarine induced HO-1 expression, and that the inhibition of MMP-9 and COX-2 expression and the enzymatic activity of sanguinarine were abrogated by siRNA-mediated knockdown of HO-1 expression. sanguinarine 26-38 heme oxygenase 1 Homo sapiens 207-211 24220687-10 2014 We also demonstrated that sanguinarine induced HO-1 expression, and that the inhibition of MMP-9 and COX-2 expression and the enzymatic activity of sanguinarine were abrogated by siRNA-mediated knockdown of HO-1 expression. sanguinarine 148-160 heme oxygenase 1 Homo sapiens 207-211 24220687-11 2014 Thus, knockdown of endogenous HO-1 decreased TPA-induced cell invasion. Tetradecanoylphorbol Acetate 45-48 heme oxygenase 1 Homo sapiens 30-34 24220687-12 2014 Overall, the results of the present study demonstrate that HO-1 plays a pivotal role in the anti-invasive response of sanguinarine in TPA-stimulated breast cancer cells. sanguinarine 118-130 heme oxygenase 1 Homo sapiens 59-63 25136403-5 2014 HO-1 isoform has been extensively studied mainly by its ability to respond to cellular stresses such as hemin, nitric oxide donors, oxidative damage, hypoxia, hyperthermia, and heavy metals, between others. Hemin 104-109 heme oxygenase 1 Homo sapiens 0-4 25136403-5 2014 HO-1 isoform has been extensively studied mainly by its ability to respond to cellular stresses such as hemin, nitric oxide donors, oxidative damage, hypoxia, hyperthermia, and heavy metals, between others. Nitric Oxide 111-123 heme oxygenase 1 Homo sapiens 0-4 24220687-12 2014 Overall, the results of the present study demonstrate that HO-1 plays a pivotal role in the anti-invasive response of sanguinarine in TPA-stimulated breast cancer cells. Tetradecanoylphorbol Acetate 134-137 heme oxygenase 1 Homo sapiens 59-63 24854244-7 2014 Similarly, lysophosphatidic acid (LPA) induced TGF-beta expression and decreased HO-1 expression in human lung fibroblasts. lysophosphatidic acid 11-32 heme oxygenase 1 Homo sapiens 81-85 24854244-7 2014 Similarly, lysophosphatidic acid (LPA) induced TGF-beta expression and decreased HO-1 expression in human lung fibroblasts. lysophosphatidic acid 34-37 heme oxygenase 1 Homo sapiens 81-85 24145726-2 2013 Of interest, patients with breast cancer have been noted to have an increase in endogenous carbon monoxide production via upregulation of heme oxygenase-1 activity. Carbon Monoxide 91-106 heme oxygenase 1 Homo sapiens 138-154 25462065-8 2014 HO-1 mRNA expression was measured as a downstream marker of response to oxidative stress induced by Zn(2+) exposure. Zinc 100-103 heme oxygenase 1 Homo sapiens 0-4 25462065-10 2014 Compartment-directed catalase expression blunted Zn(2+)-induced elevations in cytosolic EGSH and the increased expression of HO-1 mRNA levels. Zinc 49-51 heme oxygenase 1 Homo sapiens 125-129 24211270-1 2014 Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Heme 96-100 heme oxygenase 1 Homo sapiens 0-21 24211270-1 2014 Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Bilirubin 106-115 heme oxygenase 1 Homo sapiens 0-21 24211270-1 2014 Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Ferric enterobactin ion 117-123 heme oxygenase 1 Homo sapiens 0-21 24211270-1 2014 Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Carbon Monoxide 133-148 heme oxygenase 1 Homo sapiens 0-21 24211270-1 2014 Heme oxygenase (HO)-1 is an oxidative stress-response enzyme which catalyzes the degradation of heme into bilirubin, ferric ion, and carbon monoxide (CO). Carbon Monoxide 150-152 heme oxygenase 1 Homo sapiens 0-21 24428990-4 2014 The activities of the heme oxygenase-1 (HO-1) and NAD (P) H: quinone oxidoreductase 1 (NQO-1) in cells and the superoxide dismutase (SOD) and catalase (CAT) in supernatant, as well as malondialdehyde (MDA) content, were measured by enzyme-linked immunosorbent assay. Malondialdehyde 184-199 heme oxygenase 1 Homo sapiens 40-44 24428990-4 2014 The activities of the heme oxygenase-1 (HO-1) and NAD (P) H: quinone oxidoreductase 1 (NQO-1) in cells and the superoxide dismutase (SOD) and catalase (CAT) in supernatant, as well as malondialdehyde (MDA) content, were measured by enzyme-linked immunosorbent assay. Malondialdehyde 201-204 heme oxygenase 1 Homo sapiens 40-44 25593981-5 2014 Within few hours of application, disulfiram caused irreversible cell damage associated with pronounced induction of the inducible heat shock proteins HSP70, HSP40, and HSP32. Disulfiram 33-43 heme oxygenase 1 Homo sapiens 168-173 24564601-2 2014 Although they do not share the same mechanisms, membrane glycosphingolipids (GSL) and the antioxidant enzyme heme oxygenase-1 (HMOX1) both act against the pro-oxidative effect of TBA. tba 179-182 heme oxygenase 1 Homo sapiens 109-125 24564601-2 2014 Although they do not share the same mechanisms, membrane glycosphingolipids (GSL) and the antioxidant enzyme heme oxygenase-1 (HMOX1) both act against the pro-oxidative effect of TBA. tba 179-182 heme oxygenase 1 Homo sapiens 127-132 24145726-9 2013 Future investigation of the role played by heme oxygenase-1-derived carbon monoxide in the pathogenesis of breast cancer-related thrombophilia is warranted. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 43-59 23954820-5 2013 Moreover, treatments of PQ strongly inhibited the expression of Nrf2 and the downstream effectors, HO1 and NQO1. Paraquat 24-26 heme oxygenase 1 Homo sapiens 99-102 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Heme 84-88 heme oxygenase 1 Homo sapiens 0-16 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Heme 84-88 heme oxygenase 1 Homo sapiens 18-23 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 0-16 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 18-23 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 0-16 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 18-23 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Iron 130-134 heme oxygenase 1 Homo sapiens 0-16 23939757-1 2013 Heme oxygenase-1 (HMOX1) is a ubiquitously expressed inducible enzyme that degrades heme to carbon monoxide, biliverdin, and free iron ions. Iron 130-134 heme oxygenase 1 Homo sapiens 18-23 23418094-0 2013 Caffeoylserotonin protects human keratinocyte HaCaT cells against H2 O2 -induced oxidative stress and apoptosis through upregulation of HO-1 expression via activation of the PI3K/Akt/Nrf2 pathway. N-caffeoylserotonin 0-17 heme oxygenase 1 Homo sapiens 136-140 23418094-0 2013 Caffeoylserotonin protects human keratinocyte HaCaT cells against H2 O2 -induced oxidative stress and apoptosis through upregulation of HO-1 expression via activation of the PI3K/Akt/Nrf2 pathway. Hydrogen Peroxide 66-71 heme oxygenase 1 Homo sapiens 136-140 23418094-10 2013 These results demonstrate that CaS protects against oxidative stress-induced keratinocyte cell death in part through the activation of Nrf2-mediated HO-1 induction via the PI3K/Akt and/or PKC pathways, but not MAPK signaling. cas 31-34 heme oxygenase 1 Homo sapiens 149-153 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Sunitinib 86-95 heme oxygenase 1 Homo sapiens 45-61 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Sunitinib 86-95 heme oxygenase 1 Homo sapiens 63-67 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Imatinib Mesylate 100-108 heme oxygenase 1 Homo sapiens 45-61 24167056-5 2013 Among the differentially expressed proteins, heme oxygenase-1 (HO-1) was found in the sunitinib and imatinib mesylate treatment groups, which possibly acts as a specific target for synthetic lethality in combinational treatment. Imatinib Mesylate 100-108 heme oxygenase 1 Homo sapiens 63-67 24260155-7 2013 The pretreatment of cells with specific ER inhibitors or PI3K/Akt (ICI182,780 and LY294002) increased Nrf2, HO-1, and NQO1 protein, impaired nuclear translocation of HA-Nrf2, and decreased ARE-luciferase activity. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 82-90 heme oxygenase 1 Homo sapiens 108-112 23580157-7 2013 Fluticasone propionate did not modify intracellular ROS production, GSH and HDCA-2 but reduced Nrf2 and HO-1 in CSE-stimulated 16-HBE. Fluticasone 0-22 heme oxygenase 1 Homo sapiens 104-108 23580157-8 2013 Carbocysteine reduced ROS production and increased GSH, HO-1, Nrf2 and HDAC-2 nuclear expression/activity in CSE-stimulated cells and was more effective than fluticasone propionate in modulating the CSE-mediated effects. Carbocysteine 0-13 heme oxygenase 1 Homo sapiens 56-60 24121491-2 2013 Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 73-89 24121491-2 2013 Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 91-95 24121491-2 2013 Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 73-89 24121491-2 2013 Carbon monoxide (CO), one of the products of the cytoprotective molecule heme oxygenase-1 (HO-1) in cancer cells, has been implicated in carcinogenesis and therapeutic resistance. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 91-95 23877636-1 2013 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in bilirubin metabolism, and its genetic variant may modulate hyperbilirubinemia risk in neonates. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 0-16 23877636-1 2013 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in bilirubin metabolism, and its genetic variant may modulate hyperbilirubinemia risk in neonates. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 18-22 24772975-6 2013 The Grp78, MTJ1 and HMOX1 gene expression increased with the administration of 1 microM dex. Dexamethasone 88-91 heme oxygenase 1 Homo sapiens 20-25 24772975-9 2013 The increase in Grp78, Grp94, HMOX1 and MTJ1 gene expression was greater in groups in which dex was administered in combination with PBA than in groups in which dex was administered alone. Dexamethasone 92-95 heme oxygenase 1 Homo sapiens 30-35 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Heme 36-40 heme oxygenase 1 Homo sapiens 0-16 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Heme 36-40 heme oxygenase 1 Homo sapiens 18-22 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Carbon Monoxide 44-59 heme oxygenase 1 Homo sapiens 0-16 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Carbon Monoxide 44-59 heme oxygenase 1 Homo sapiens 18-22 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. ammonium ferrous sulfate 61-68 heme oxygenase 1 Homo sapiens 0-16 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. ammonium ferrous sulfate 61-68 heme oxygenase 1 Homo sapiens 18-22 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Biliverdine 78-88 heme oxygenase 1 Homo sapiens 0-16 24323422-7 2013 Heme oxygenase-1 (HO-1) metabolizes heme to carbon monoxide, ferrous ion, and biliverdin. Biliverdine 78-88 heme oxygenase 1 Homo sapiens 18-22 24323422-8 2013 CO activates cGMP to promote vasodilation, and biliverdin is converted to bilirubin which can serve as an anti-oxidant, both of which may contribute to the reported protective role of HO-1 in cerebral ischemia and subarachnoid hemorrhage. Biliverdine 47-57 heme oxygenase 1 Homo sapiens 184-188 24323422-8 2013 CO activates cGMP to promote vasodilation, and biliverdin is converted to bilirubin which can serve as an anti-oxidant, both of which may contribute to the reported protective role of HO-1 in cerebral ischemia and subarachnoid hemorrhage. Bilirubin 74-83 heme oxygenase 1 Homo sapiens 184-188 24323422-9 2013 However, ferrous ion can react with hydrogen peroxide to produce pro-oxidant hydroxyl radicals which may explain the harmful role of HO-1 in intracerebral hemorrhage. ammonium ferrous sulfate 9-20 heme oxygenase 1 Homo sapiens 133-137 24323422-9 2013 However, ferrous ion can react with hydrogen peroxide to produce pro-oxidant hydroxyl radicals which may explain the harmful role of HO-1 in intracerebral hemorrhage. Hydrogen Peroxide 36-53 heme oxygenase 1 Homo sapiens 133-137 24323422-9 2013 However, ferrous ion can react with hydrogen peroxide to produce pro-oxidant hydroxyl radicals which may explain the harmful role of HO-1 in intracerebral hemorrhage. Hydroxyl Radical 77-94 heme oxygenase 1 Homo sapiens 133-137 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Heme 99-103 heme oxygenase 1 Homo sapiens 0-16 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Heme 99-103 heme oxygenase 1 Homo sapiens 18-22 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 0-16 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 18-22 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Carbon Monoxide 126-129 heme oxygenase 1 Homo sapiens 0-16 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Carbon Monoxide 126-129 heme oxygenase 1 Homo sapiens 18-22 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. ammonium ferrous sulfate 131-137 heme oxygenase 1 Homo sapiens 0-16 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. ammonium ferrous sulfate 131-137 heme oxygenase 1 Homo sapiens 18-22 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 0-16 23852701-1 2013 Heme oxygenase-1 (HO-1) inhibits immune responses and inflammatory reactions via the catabolism of heme into carbon monoxide (CO), Fe(2+) , and biliverdin. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 18-22 23836589-8 2013 Both heme oxygenase 1 (HO-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) knockdown attenuated the isothiocyanate inhibition of oxLDL-induced ROS production, kappaB-reporter activity, and adhesion molecule expression. isothiocyanic acid 109-123 heme oxygenase 1 Homo sapiens 5-21 23426167-7 2013 Inhibition of HO-1 increases the toxicity of ATO, but has no effect on Dar-induced apoptosis. Arsenic Trioxide 45-48 heme oxygenase 1 Homo sapiens 14-18 24201221-3 2013 HMOX1 is the rate-limiting enzyme of heme degradation, and the heme breakdown products, CO, Fe, and bilirubin, are considered to be biologically active metabolites with direct or indirect antioxidant and anti-inflammatory properties. Heme 37-41 heme oxygenase 1 Homo sapiens 0-5 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. 4-hydroxy-2-nonenal 58-63 heme oxygenase 1 Homo sapiens 179-183 24179613-2 2013 HO-1, a stress-responsive enzyme that catabolizes heme into carbon monoxide (CO), biliverdin and iron, has previously been shown to protect grafts from ischemia/reperfusion and rejection. Heme 50-54 heme oxygenase 1 Homo sapiens 0-4 24179613-2 2013 HO-1, a stress-responsive enzyme that catabolizes heme into carbon monoxide (CO), biliverdin and iron, has previously been shown to protect grafts from ischemia/reperfusion and rejection. Carbon Monoxide 60-75 heme oxygenase 1 Homo sapiens 0-4 24179613-2 2013 HO-1, a stress-responsive enzyme that catabolizes heme into carbon monoxide (CO), biliverdin and iron, has previously been shown to protect grafts from ischemia/reperfusion and rejection. Carbon Monoxide 77-79 heme oxygenase 1 Homo sapiens 0-4 24179613-2 2013 HO-1, a stress-responsive enzyme that catabolizes heme into carbon monoxide (CO), biliverdin and iron, has previously been shown to protect grafts from ischemia/reperfusion and rejection. Biliverdine 82-92 heme oxygenase 1 Homo sapiens 0-4 24179613-2 2013 HO-1, a stress-responsive enzyme that catabolizes heme into carbon monoxide (CO), biliverdin and iron, has previously been shown to protect grafts from ischemia/reperfusion and rejection. Iron 97-101 heme oxygenase 1 Homo sapiens 0-4 24179613-5 2013 More specifically, the CO derived from HO-1-mediated heme catabolism has been shown to be involved in the regulation of inflammation; furthermore, administration of low concentrations of exogenous CO has a protective effect against inflammation. Heme 53-57 heme oxygenase 1 Homo sapiens 39-43 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. nitrolipid 97-107 heme oxygenase 1 Homo sapiens 179-183 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. 4-hydroxy-2-nonenal 255-260 heme oxygenase 1 Homo sapiens 179-183 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. 10-nitro-oleic acid 67-84 heme oxygenase 1 Homo sapiens 179-183 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. 6H 264-266 heme oxygenase 1 Homo sapiens 179-183 24075775-0 2013 Docosahexaenoic acid (DHA)-induced heme oxygenase-1 attenuates cytotoxic effects of DHA in vascular smooth muscle cells. Docosahexaenoic Acids 0-20 heme oxygenase 1 Homo sapiens 35-51 23845594-6 2013 In human corneal epithelial cells in culture, addition of 4-HNE or 9-nitrooleic acid, a reactive nitrolipid formed during nitrosative stress, caused a time-dependent induction of HO-1 mRNA and protein; maximal responses were evident after 10h with 30 muM 4-HNE or 6h with 10 muM 9-nitrooleic acid. 10-nitro-oleic acid 279-296 heme oxygenase 1 Homo sapiens 179-183 24052896-3 2013 A known regulator of the oxidative stress response is the heat shock protein, heme-oxygenase 1 (HO-1), which is induced by cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 123-147 heme oxygenase 1 Homo sapiens 78-94 24052896-3 2013 A known regulator of the oxidative stress response is the heat shock protein, heme-oxygenase 1 (HO-1), which is induced by cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 149-153 heme oxygenase 1 Homo sapiens 78-94 23816986-0 2013 The activation of HO-1/Nrf-2 contributes to the protective effects of diallyl disulfide (DADS) against ethanol-induced oxidative stress. diallyl disulfide 70-87 heme oxygenase 1 Homo sapiens 18-22 23816986-0 2013 The activation of HO-1/Nrf-2 contributes to the protective effects of diallyl disulfide (DADS) against ethanol-induced oxidative stress. diallyl disulfide 89-93 heme oxygenase 1 Homo sapiens 18-22 23816986-0 2013 The activation of HO-1/Nrf-2 contributes to the protective effects of diallyl disulfide (DADS) against ethanol-induced oxidative stress. Ethanol 103-110 heme oxygenase 1 Homo sapiens 18-22 23816986-12 2013 CONCLUSION: These results demonstrate that DADS could induce the activation of HO-1/Nrf-2 pathway, which may contribute to the protective effects of DADS against ethanol-induced liver injury. diallyl disulfide 43-47 heme oxygenase 1 Homo sapiens 79-83 23816986-12 2013 CONCLUSION: These results demonstrate that DADS could induce the activation of HO-1/Nrf-2 pathway, which may contribute to the protective effects of DADS against ethanol-induced liver injury. diallyl disulfide 149-153 heme oxygenase 1 Homo sapiens 79-83 23816986-12 2013 CONCLUSION: These results demonstrate that DADS could induce the activation of HO-1/Nrf-2 pathway, which may contribute to the protective effects of DADS against ethanol-induced liver injury. Ethanol 162-169 heme oxygenase 1 Homo sapiens 79-83 24075775-0 2013 Docosahexaenoic acid (DHA)-induced heme oxygenase-1 attenuates cytotoxic effects of DHA in vascular smooth muscle cells. Docosahexaenoic Acids 22-25 heme oxygenase 1 Homo sapiens 35-51 24075775-0 2013 Docosahexaenoic acid (DHA)-induced heme oxygenase-1 attenuates cytotoxic effects of DHA in vascular smooth muscle cells. Docosahexaenoic Acids 84-87 heme oxygenase 1 Homo sapiens 35-51 24075775-3 2013 In the present study we examined the impact of heme oxygenase (HO)-1, induced by DHA, on DHA-induced oxidative stress, UPR, cell proliferation and apoptosis in hPASMC. Docosahexaenoic Acids 81-84 heme oxygenase 1 Homo sapiens 47-68 24075775-3 2013 In the present study we examined the impact of heme oxygenase (HO)-1, induced by DHA, on DHA-induced oxidative stress, UPR, cell proliferation and apoptosis in hPASMC. Docosahexaenoic Acids 89-92 heme oxygenase 1 Homo sapiens 47-68 23932761-6 2013 Consistent with this view, high expression alleles of the major enzymatic source of bilirubin, heme oxygenase-1 (HO-1), do associate with decreased vascular risk in the majority of studies that have addressed this issue, and increased plasma bilirubin has been reported in carriers of these alleles. Bilirubin 84-93 heme oxygenase 1 Homo sapiens 95-111 23347118-4 2013 EXPERIMENTAL APPROACH: The aim of this study was to analyse the effects of AEA or FAAH inhibition by the URB597 inhibitor or FAAH/siRNA on the activation of Nrf2-ARE signalling pathway and heme oxygenase-1 (HO-1) induction and transcription. aea 75-78 heme oxygenase 1 Homo sapiens 189-205 23347118-4 2013 EXPERIMENTAL APPROACH: The aim of this study was to analyse the effects of AEA or FAAH inhibition by the URB597 inhibitor or FAAH/siRNA on the activation of Nrf2-ARE signalling pathway and heme oxygenase-1 (HO-1) induction and transcription. aea 75-78 heme oxygenase 1 Homo sapiens 207-211 23347118-7 2013 We found that inhibition of FAAH by the URB597 inhibitor induced antioxidant HO-1 in breast cancer cells and MCF-10A cells. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 40-46 heme oxygenase 1 Homo sapiens 77-81 23347118-9 2013 Furthermore, URB597, AEA and siRNA-FAAH treatments induced the nuclear translocation of Nrf2, while siRNA-Nrf2 treatment and Keap1 expression blocked AEA, URB597 and si-FAAH from activation of ARE reporter and HO-1 induction. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 13-19 heme oxygenase 1 Homo sapiens 210-214 23347118-9 2013 Furthermore, URB597, AEA and siRNA-FAAH treatments induced the nuclear translocation of Nrf2, while siRNA-Nrf2 treatment and Keap1 expression blocked AEA, URB597 and si-FAAH from activation of ARE reporter and HO-1 induction. aea 150-153 heme oxygenase 1 Homo sapiens 210-214 23347118-9 2013 Furthermore, URB597, AEA and siRNA-FAAH treatments induced the nuclear translocation of Nrf2, while siRNA-Nrf2 treatment and Keap1 expression blocked AEA, URB597 and si-FAAH from activation of ARE reporter and HO-1 induction. cyclohexyl carbamic acid 3'-carbamoylbiphenyl-3-yl ester 155-161 heme oxygenase 1 Homo sapiens 210-214 23347118-11 2013 CONCLUSIONS AND IMPLICATIONS: These data uncovered a novel mechanism by which inhibition of FAAH or exposure to AEA induced HO-1 transcripts and implicating AEA and FAAH as direct modifiers in signalling mediated activation of Nrf2-HO-1 pathway, independent of cannabinoid receptors. aea 112-115 heme oxygenase 1 Homo sapiens 124-128 23347118-11 2013 CONCLUSIONS AND IMPLICATIONS: These data uncovered a novel mechanism by which inhibition of FAAH or exposure to AEA induced HO-1 transcripts and implicating AEA and FAAH as direct modifiers in signalling mediated activation of Nrf2-HO-1 pathway, independent of cannabinoid receptors. aea 112-115 heme oxygenase 1 Homo sapiens 232-236 23347118-11 2013 CONCLUSIONS AND IMPLICATIONS: These data uncovered a novel mechanism by which inhibition of FAAH or exposure to AEA induced HO-1 transcripts and implicating AEA and FAAH as direct modifiers in signalling mediated activation of Nrf2-HO-1 pathway, independent of cannabinoid receptors. aea 157-160 heme oxygenase 1 Homo sapiens 124-128 23347118-11 2013 CONCLUSIONS AND IMPLICATIONS: These data uncovered a novel mechanism by which inhibition of FAAH or exposure to AEA induced HO-1 transcripts and implicating AEA and FAAH as direct modifiers in signalling mediated activation of Nrf2-HO-1 pathway, independent of cannabinoid receptors. aea 157-160 heme oxygenase 1 Homo sapiens 232-236 23813560-3 2013 This study assessed the association between the length of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene microsatellite promoter and cardiovascular events and mortality among hemodialysis patients. guanosine thymidine dinucleotide 58-90 heme oxygenase 1 Homo sapiens 106-122 23813560-5 2013 The allelic frequencies of the length of guanosine thymidine dinucleotide repeats (the S allele represents shorter [<27] repeats, and the L allele represents longer [>= 27] repeats) in the heme oxygenase-1 gene promoter were analyzed in 1080 unrelated chronic hemodialysis patients and 365 healthy controls for distribution comparison. guanosine thymidine dinucleotide 41-73 heme oxygenase 1 Homo sapiens 195-211 23813560-12 2013 CONCLUSIONS: Chronic hemodialysis patients with longer lengths of guanosine thymidine dinucleotide repeats in the heme oxygenase-1 gene promoter exhibit higher inflammation and oxidative stress. guanosine thymidine dinucleotide 66-98 heme oxygenase 1 Homo sapiens 114-130 22593043-3 2013 We found that the endogenous expression of Nrf2 as well as its target proteins heme oxygenase-1 (HO-1), NADP(H):quinone oxidoreductase (NQO) in MCF-7/DOX cells was higher than that in MCF-7 cells. Doxorubicin 150-153 heme oxygenase 1 Homo sapiens 79-95 22593043-3 2013 We found that the endogenous expression of Nrf2 as well as its target proteins heme oxygenase-1 (HO-1), NADP(H):quinone oxidoreductase (NQO) in MCF-7/DOX cells was higher than that in MCF-7 cells. Doxorubicin 150-153 heme oxygenase 1 Homo sapiens 97-101 22593043-4 2013 Tert-butylhydroquinone treatment increased the expression Nrf2, HO-1, and NQO-1, and enhanced resistance of MCF-7 cells to DOX. 2-tert-butylhydroquinone 0-22 heme oxygenase 1 Homo sapiens 64-68 23872045-4 2013 The mRNA expression levels of detoxifying, NQO1, HO-1, GSTA1, MT-1, and HSP70, were significantly decreased in all heavy metal-exposed group as compared to control group. Metals 121-126 heme oxygenase 1 Homo sapiens 49-53 23759444-4 2013 Xenon exposure enhanced the expression of heat-shock protein 70 (HSP-70) and heme oxygenase 1 (HO-1) and promoted cell survival after hypothermia-hypoxia insult in human proximal tubular (HK-2) cells, which was abolished by HSP-70 or HO-1 siRNA. Xenon 0-5 heme oxygenase 1 Homo sapiens 77-93 23759444-4 2013 Xenon exposure enhanced the expression of heat-shock protein 70 (HSP-70) and heme oxygenase 1 (HO-1) and promoted cell survival after hypothermia-hypoxia insult in human proximal tubular (HK-2) cells, which was abolished by HSP-70 or HO-1 siRNA. Xenon 0-5 heme oxygenase 1 Homo sapiens 95-99 23759444-4 2013 Xenon exposure enhanced the expression of heat-shock protein 70 (HSP-70) and heme oxygenase 1 (HO-1) and promoted cell survival after hypothermia-hypoxia insult in human proximal tubular (HK-2) cells, which was abolished by HSP-70 or HO-1 siRNA. Xenon 0-5 heme oxygenase 1 Homo sapiens 234-238 23850497-1 2013 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. Heme 111-115 heme oxygenase 1 Homo sapiens 0-16 23850497-1 2013 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. Heme 111-115 heme oxygenase 1 Homo sapiens 18-22 23850497-1 2013 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. Iron 137-141 heme oxygenase 1 Homo sapiens 0-16 23850497-1 2013 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme that is induced by intraplaque hemorrhage and degrades free heme and releases ferrous iron, which is rapidly sequestered by ferritin. Iron 137-141 heme oxygenase 1 Homo sapiens 18-22 23932761-6 2013 Consistent with this view, high expression alleles of the major enzymatic source of bilirubin, heme oxygenase-1 (HO-1), do associate with decreased vascular risk in the majority of studies that have addressed this issue, and increased plasma bilirubin has been reported in carriers of these alleles. Bilirubin 242-251 heme oxygenase 1 Homo sapiens 95-111 24003391-12 2013 CONCLUSIONS: AKAP plays an important role in PGE2 regulation of COX-2 expression, and the suppression of HO-1 by PGE2-cAMP-PKA-AKAP signaling helps potentiate the LPS-induced COX-2 expression through a positive feedback loop in RAW 264.7 macrophages. Dinoprostone 113-117 heme oxygenase 1 Homo sapiens 105-109 23824679-0 2013 Role of HO-1 in the arsenite-induced neurotoxicity in primary cultured cortical neurons. arsenite 20-28 heme oxygenase 1 Homo sapiens 8-12 23824679-1 2013 In the present study, the role of heme oxygenase (HO)-1 in sodium arsenite (arsenite)-induced neurotoxicity was investigated using primary cultured cortical neurons. sodium arsenite 59-74 heme oxygenase 1 Homo sapiens 34-55 23824679-1 2013 In the present study, the role of heme oxygenase (HO)-1 in sodium arsenite (arsenite)-induced neurotoxicity was investigated using primary cultured cortical neurons. arsenite 66-74 heme oxygenase 1 Homo sapiens 34-55 23824679-5 2013 Western blot assay showed that arsenite significantly increased HO-1 levels, a redox-regulated protein. arsenite 31-39 heme oxygenase 1 Homo sapiens 64-68 23824679-6 2013 Co-incubation with glutathione (10 mM) attenuated arsenite-induced HO-1 elevation and caspase 3 activation, suggesting that oxidative stress is involved in the arsenite-induced neurotoxicity. Glutathione 19-30 heme oxygenase 1 Homo sapiens 67-71 23824679-6 2013 Co-incubation with glutathione (10 mM) attenuated arsenite-induced HO-1 elevation and caspase 3 activation, suggesting that oxidative stress is involved in the arsenite-induced neurotoxicity. arsenite 50-58 heme oxygenase 1 Homo sapiens 67-71 23824679-7 2013 The neurotoxic effects of inorganic arsenics were compared; arsenite was more potent than arsenate in inducing HO-1 expression and caspase 3 activation. arsenite 60-68 heme oxygenase 1 Homo sapiens 111-115 23824679-7 2013 The neurotoxic effects of inorganic arsenics were compared; arsenite was more potent than arsenate in inducing HO-1 expression and caspase 3 activation. arsenic acid 90-98 heme oxygenase 1 Homo sapiens 111-115 23824679-9 2013 HO-1 siRNA transfection was employed to prevent arsenite-induced HO-1 elevation. arsenite 48-56 heme oxygenase 1 Homo sapiens 0-4 24003391-5 2013 METHODS: We explored the involvement of HO-1 on PGE2 regulation of LPS-induced COX-2 expression in RAW 264.7 macrophages. Dinoprostone 48-52 heme oxygenase 1 Homo sapiens 40-44 24003391-10 2013 In addition, HO-1 induction by the HO activator copper protoporphyrin suppressed LPS-induced COX-2 expression, which was restored by the addition of exogenous PGE2. Copper 48-54 heme oxygenase 1 Homo sapiens 13-17 24003391-10 2013 In addition, HO-1 induction by the HO activator copper protoporphyrin suppressed LPS-induced COX-2 expression, which was restored by the addition of exogenous PGE2. protoporphyrin IX 55-69 heme oxygenase 1 Homo sapiens 13-17 24003391-10 2013 In addition, HO-1 induction by the HO activator copper protoporphyrin suppressed LPS-induced COX-2 expression, which was restored by the addition of exogenous PGE2. Dinoprostone 159-163 heme oxygenase 1 Homo sapiens 13-17 23824679-9 2013 HO-1 siRNA transfection was employed to prevent arsenite-induced HO-1 elevation. arsenite 48-56 heme oxygenase 1 Homo sapiens 65-69 23824679-10 2013 At the same time, arsenite-induced caspase 3 activation and neuronal death were attenuated in the HO-1 siRNA-transfected cells. arsenite 18-26 heme oxygenase 1 Homo sapiens 98-102 23824679-11 2013 Taken together, HO-1 appears to be neuroprotective in the arsenite-induced neurotoxicity in primary cultured cortical neurons. arsenite 58-66 heme oxygenase 1 Homo sapiens 16-20 23824679-12 2013 In addition to antioxidants, HO-1 elevation may be a neuroprotective strategy for arsenite-induced neurotoxicity. arsenite 82-90 heme oxygenase 1 Homo sapiens 29-33 24046187-0 2013 S-adenosyl methionine prevents endothelial dysfunction by inducing heme oxygenase-1 in vascular endothelial cells. Sulfur 0-1 heme oxygenase 1 Homo sapiens 67-83 24046187-0 2013 S-adenosyl methionine prevents endothelial dysfunction by inducing heme oxygenase-1 in vascular endothelial cells. S-Adenosylmethionine 2-21 heme oxygenase 1 Homo sapiens 67-83 24003391-12 2013 CONCLUSIONS: AKAP plays an important role in PGE2 regulation of COX-2 expression, and the suppression of HO-1 by PGE2-cAMP-PKA-AKAP signaling helps potentiate the LPS-induced COX-2 expression through a positive feedback loop in RAW 264.7 macrophages. Cyclic AMP 118-122 heme oxygenase 1 Homo sapiens 105-109 23617628-6 2013 Carbon monoxide was a mediator of HMOX1 effects on proliferation, migration, and miR-378 expression. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 34-39 23867390-1 2013 A novel series of aryloxyalkyl derivatives of imidazole and 1,2,4-triazole, 17-31, was designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). 1,2,4-triazole 60-74 heme oxygenase 1 Homo sapiens 129-145 23867390-0 2013 Evaluation of novel aryloxyalkyl derivatives of imidazole and 1,2,4-triazole as heme oxygenase-1 (HO-1) inhibitors and their antitumor properties. imidazole 48-57 heme oxygenase 1 Homo sapiens 80-96 23867390-1 2013 A novel series of aryloxyalkyl derivatives of imidazole and 1,2,4-triazole, 17-31, was designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). 1,2,4-triazole 60-74 heme oxygenase 1 Homo sapiens 147-151 23867390-0 2013 Evaluation of novel aryloxyalkyl derivatives of imidazole and 1,2,4-triazole as heme oxygenase-1 (HO-1) inhibitors and their antitumor properties. imidazole 48-57 heme oxygenase 1 Homo sapiens 98-102 23867390-2 2013 Some of these compounds were found to be good inhibitors of HO-1, in particular those carrying an imidazole moiety as azolyl group and a 3-bromo or 4-iodophenyl as aryl moiety. imidazole 98-107 heme oxygenase 1 Homo sapiens 60-64 23867390-0 2013 Evaluation of novel aryloxyalkyl derivatives of imidazole and 1,2,4-triazole as heme oxygenase-1 (HO-1) inhibitors and their antitumor properties. 1,2,4-triazole 62-76 heme oxygenase 1 Homo sapiens 80-96 22609818-0 2013 Genetic polymorphisms of HO-1 and COX-1 are associated with aspirin resistance defined by light transmittance aggregation in Chinese Han patients. Aspirin 60-67 heme oxygenase 1 Homo sapiens 25-29 23867390-0 2013 Evaluation of novel aryloxyalkyl derivatives of imidazole and 1,2,4-triazole as heme oxygenase-1 (HO-1) inhibitors and their antitumor properties. 1,2,4-triazole 62-76 heme oxygenase 1 Homo sapiens 98-102 23867390-1 2013 A novel series of aryloxyalkyl derivatives of imidazole and 1,2,4-triazole, 17-31, was designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). imidazole 46-55 heme oxygenase 1 Homo sapiens 129-145 23867390-1 2013 A novel series of aryloxyalkyl derivatives of imidazole and 1,2,4-triazole, 17-31, was designed and synthesized as inhibitors of heme oxygenase-1 (HO-1) and heme oxygenase-2 (HO-2). imidazole 46-55 heme oxygenase 1 Homo sapiens 147-151 23817544-2 2013 Of interest, thyroid cancer cell lines can have upregulation of the carbon monoxide-producing enzyme, hemeoxygenase-1. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 102-117 23817544-5 2013 This initial observation serves as a rationale to further investigate the role played by hemeoxygenase-1 upregulation in the setting of cancers associated with increased endogenous carbon monoxide production. Carbon Monoxide 181-196 heme oxygenase 1 Homo sapiens 89-104 22609818-1 2013 BACKGROUND: Cyclooxygenase 1 (COX-1), COX-2, and HO-1 are involved in the process of aspirin"s effect. Aspirin 85-92 heme oxygenase 1 Homo sapiens 49-53 22609818-10 2013 HO-1 rs2071746 (-413A>T) is associated with AR. Argon 47-49 heme oxygenase 1 Homo sapiens 0-4 25337552-3 2013 In the present study, the cytoprotective effects of morin (3,5,7,2",4"-pentahydroxyflavone), in terms of HO-1 enzyme, against the oxidative stress and its involved mechanisms was elucidated. morin 59-90 heme oxygenase 1 Homo sapiens 105-109 23712098-0 2013 Lucidone protects human skin keratinocytes against free radical-induced oxidative damage and inflammation through the up-regulation of HO-1/Nrf2 antioxidant genes and down-regulation of NF-kappaB signaling pathway. lucidone 0-8 heme oxygenase 1 Homo sapiens 135-139 23729024-0 2013 Effectiveness of miltefosine treatment in targeting anti-leishmanial HO-1/Nrf-2-mediated oxidative responses in visceral leishmaniasis patients. miltefosine 17-28 heme oxygenase 1 Homo sapiens 69-73 23729024-3 2013 We aimed to investigate the role of miltefosine in regulating HO-1/Nrf-2-mediated oxidative responses in visceral leishmaniasis (VL) patients in vivo and in vitro. miltefosine 36-47 heme oxygenase 1 Homo sapiens 62-66 23729024-7 2013 The in vitro HO-1/Nrf-2-dependent anti-leishmanial effect of miltefosine was assessed by the use of specific inhibitors/inducers and subsequent microscopic measurement of parasite killing and Th1/Th2 cytokine regulation by ELISA. miltefosine 61-72 heme oxygenase 1 Homo sapiens 13-17 23729024-8 2013 RESULTS: Increased levels of transcript and serum HO-1, Nrf-2 nuclear translocation, serum bilirubin, GPx and SOD activity in untreated VL patients were reversed after miltefosine chemotherapy. miltefosine 168-179 heme oxygenase 1 Homo sapiens 50-54 23729024-9 2013 The effectiveness of miltefosine for positive induction of ROS via NADPH correlated with a decrease in HO-1/ERK/Nrf-2-dependent parasite load. miltefosine 21-32 heme oxygenase 1 Homo sapiens 103-107 23729024-9 2013 The effectiveness of miltefosine for positive induction of ROS via NADPH correlated with a decrease in HO-1/ERK/Nrf-2-dependent parasite load. Reactive Oxygen Species 59-62 heme oxygenase 1 Homo sapiens 103-107 23729024-9 2013 The effectiveness of miltefosine for positive induction of ROS via NADPH correlated with a decrease in HO-1/ERK/Nrf-2-dependent parasite load. NADP 67-72 heme oxygenase 1 Homo sapiens 103-107 23729024-10 2013 Furthermore, HO-1 blockade by miltefosine led to suppression of interleukin-10 and transforming growth factor-beta, but enhanced interleukin-12 and tumour necrosis factor-alpha production, in VL patients. miltefosine 30-41 heme oxygenase 1 Homo sapiens 13-17 23712098-0 2013 Lucidone protects human skin keratinocytes against free radical-induced oxidative damage and inflammation through the up-regulation of HO-1/Nrf2 antioxidant genes and down-regulation of NF-kappaB signaling pathway. Free Radicals 51-63 heme oxygenase 1 Homo sapiens 135-139 23712098-3 2013 Notably, the antioxidant potential of lucidone was directly correlated with the increased expression of an antioxidant gene, heme oxygenase 1 (HO-1), which was followed by the augmentation of the nuclear translocation and transcriptional activation of NF-E2-related factor-2 (Nrf2), with or without AAPH. lucidone 38-46 heme oxygenase 1 Homo sapiens 125-141 23712098-3 2013 Notably, the antioxidant potential of lucidone was directly correlated with the increased expression of an antioxidant gene, heme oxygenase 1 (HO-1), which was followed by the augmentation of the nuclear translocation and transcriptional activation of NF-E2-related factor-2 (Nrf2), with or without AAPH. lucidone 38-46 heme oxygenase 1 Homo sapiens 143-147 25337552-9 2013 HO-1 inhibitor ZnPP attenuated the protective effect of morin against H2O2-induced cytotoxicity. zinc protoporphyrin 15-19 heme oxygenase 1 Homo sapiens 0-4 25337552-9 2013 HO-1 inhibitor ZnPP attenuated the protective effect of morin against H2O2-induced cytotoxicity. Hydrogen Peroxide 70-74 heme oxygenase 1 Homo sapiens 0-4 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 23661026-0 2013 Overexpression of Smad7 suppressed ROS/MMP9-dependent collagen synthesis through regulation of heme oxygenase-1. Reactive Oxygen Species 35-38 heme oxygenase 1 Homo sapiens 95-111 23661026-6 2013 Further investigation showed that Smad7 regulated NADPH-mediated ROS production through activating Heme oxygenase-1 (HO-1). NADP 50-55 heme oxygenase 1 Homo sapiens 99-115 23661026-6 2013 Further investigation showed that Smad7 regulated NADPH-mediated ROS production through activating Heme oxygenase-1 (HO-1). NADP 50-55 heme oxygenase 1 Homo sapiens 117-121 23661026-6 2013 Further investigation showed that Smad7 regulated NADPH-mediated ROS production through activating Heme oxygenase-1 (HO-1). Reactive Oxygen Species 65-68 heme oxygenase 1 Homo sapiens 99-115 23661026-6 2013 Further investigation showed that Smad7 regulated NADPH-mediated ROS production through activating Heme oxygenase-1 (HO-1). Reactive Oxygen Species 65-68 heme oxygenase 1 Homo sapiens 117-121 23661026-8 2013 Knockdown of HO-1 in Smad7-overexpressed cardiac fibroblasts or cells pretreated with SnPP IX, a competitive inhibitor of HO-1 activity, resulted in increased productions of ROS and NADPH p47(phox), and abolished the inhibitory effects of Smad7 on MMP9 activity and collagen expression. tin protoporphyrin IX 86-93 heme oxygenase 1 Homo sapiens 13-17 23661026-8 2013 Knockdown of HO-1 in Smad7-overexpressed cardiac fibroblasts or cells pretreated with SnPP IX, a competitive inhibitor of HO-1 activity, resulted in increased productions of ROS and NADPH p47(phox), and abolished the inhibitory effects of Smad7 on MMP9 activity and collagen expression. tin protoporphyrin IX 86-93 heme oxygenase 1 Homo sapiens 122-126 23661026-8 2013 Knockdown of HO-1 in Smad7-overexpressed cardiac fibroblasts or cells pretreated with SnPP IX, a competitive inhibitor of HO-1 activity, resulted in increased productions of ROS and NADPH p47(phox), and abolished the inhibitory effects of Smad7 on MMP9 activity and collagen expression. Reactive Oxygen Species 174-177 heme oxygenase 1 Homo sapiens 13-17 23707609-9 2013 The inactivation of AMPK by siRNA, DN-AMPK or the pharmacological AMPK inhibitor compound C, revealed that metformin reduced HO-1 expression in an AMPK-independent manner. Metformin 107-116 heme oxygenase 1 Homo sapiens 125-129 23661026-8 2013 Knockdown of HO-1 in Smad7-overexpressed cardiac fibroblasts or cells pretreated with SnPP IX, a competitive inhibitor of HO-1 activity, resulted in increased productions of ROS and NADPH p47(phox), and abolished the inhibitory effects of Smad7 on MMP9 activity and collagen expression. 4-ethoxymethylene-2-phenyl-2-oxazoline-5-one 192-196 heme oxygenase 1 Homo sapiens 13-17 23707609-0 2013 Metformin inhibits heme oxygenase-1 expression in cancer cells through inactivation of Raf-ERK-Nrf2 signaling and AMPK-independent pathways. Metformin 0-9 heme oxygenase 1 Homo sapiens 19-35 23707609-4 2013 In this study, we tested the hypothesis that the anti-tumor effects of metformin are mediated by suppression of HO-1 expression in cancer cells. Metformin 71-80 heme oxygenase 1 Homo sapiens 112-116 23707609-5 2013 Our results indicate that metformin strongly suppresses HO-1 mRNA and protein expression in human hepatic carcinoma HepG2, cervical cancer HeLa, and non-small-cell lung cancer A549 cells. Metformin 26-35 heme oxygenase 1 Homo sapiens 56-60 23878198-8 2013 Overexpression of PtGR-1 in HEK293T cells promoted nitroalkene metabolism to inactive nitroalkanes, an effect that abrogated the Nrf2-dependent induction of heme oxygenase-1 expression by nitro-oleic acid. nitro-oleic acid 188-204 heme oxygenase 1 Homo sapiens 157-173 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Heme 60-64 heme oxygenase 1 Homo sapiens 207-211 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 0-16 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 18-22 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 207-211 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Iron 167-171 heme oxygenase 1 Homo sapiens 0-16 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Iron 167-171 heme oxygenase 1 Homo sapiens 18-22 23714423-2 2013 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. We show in this study that HO-1 expression is reduced in PBMCs of MS patients and that during exacerbation of the disease there is a significant downregulation of this enzyme. Iron 167-171 heme oxygenase 1 Homo sapiens 207-211 23720344-2 2013 Interestingly, the antioxidant enzyme heme oxygenase-1 (HO-1) is not present in the mitochondria despite the fact that the organelle is the site of heme synthesis and contains multiple heme proteins. Heme 38-42 heme oxygenase 1 Homo sapiens 56-60 23720344-2 2013 Interestingly, the antioxidant enzyme heme oxygenase-1 (HO-1) is not present in the mitochondria despite the fact that the organelle is the site of heme synthesis and contains multiple heme proteins. Heme 148-152 heme oxygenase 1 Homo sapiens 56-60 23720344-9 2013 Furthermore, decrement in production of tricarboxylic acid cycle intermediates following hypoxia was significantly mitigated in Mito-HO-1 cells. Tricarboxylic Acids 40-58 heme oxygenase 1 Homo sapiens 133-137 23720344-10 2013 These data suggest that specific mitochondrially targeted HO-1 under acute pathological conditions may have beneficial effects, but the selective advantage of long-term expression is constrained by a negative impact on the synthesis of heme-containing mitochondrial proteins. Heme 236-240 heme oxygenase 1 Homo sapiens 58-62 23707273-0 2013 Peroxisome proliferator-activated receptor-gamma agonist rosiglitazone attenuates inflammatory pain through the induction of heme oxygenase-1 in macrophages. Rosiglitazone 57-70 heme oxygenase 1 Homo sapiens 125-141 23615401-8 2013 HO-1 end products, such as carbon monoxide, free iron and bilirubin, suppressed the TPA-induced MMP-9 mRNA and protein expression, enzyme activity, migration and invasion in MCF-7 cells. Carbon Monoxide 27-42 heme oxygenase 1 Homo sapiens 0-4 23615401-8 2013 HO-1 end products, such as carbon monoxide, free iron and bilirubin, suppressed the TPA-induced MMP-9 mRNA and protein expression, enzyme activity, migration and invasion in MCF-7 cells. Iron 49-53 heme oxygenase 1 Homo sapiens 0-4 23615401-8 2013 HO-1 end products, such as carbon monoxide, free iron and bilirubin, suppressed the TPA-induced MMP-9 mRNA and protein expression, enzyme activity, migration and invasion in MCF-7 cells. Bilirubin 58-67 heme oxygenase 1 Homo sapiens 0-4 23615401-8 2013 HO-1 end products, such as carbon monoxide, free iron and bilirubin, suppressed the TPA-induced MMP-9 mRNA and protein expression, enzyme activity, migration and invasion in MCF-7 cells. Tetradecanoylphorbol Acetate 84-87 heme oxygenase 1 Homo sapiens 0-4 23615401-9 2013 Furthermore, TPA-induced extracellular signal-regulated kinase (ERK) 1/2 and Akt phosphorylation and the DNA binding activity of activator protein-1 (AP-1) and nuclear factor-kappa B (NF-kappaB) were attenuated by pretreatment with AP and HO-1 end products. Tetradecanoylphorbol Acetate 13-16 heme oxygenase 1 Homo sapiens 239-243 23615401-11 2013 Additionally, induction of HO-1 expression is at least partially involved in the inhibition of TPA-induced MMP-9 activation and cell migration in MCF-7 cells by AP. Tetradecanoylphorbol Acetate 95-98 heme oxygenase 1 Homo sapiens 27-31 23624303-0 2013 Immuno-spin trapping of heme-induced protein radicals: Implications for heme oxygenase-1 induction and heme degradation. Heme 24-28 heme oxygenase 1 Homo sapiens 72-88 23624303-3 2013 Heme is known to induce heme oxygenase-1(HO-1) expression, and the extent of induction depends on the ratio of albumin to heme in plasma. Heme 0-4 heme oxygenase 1 Homo sapiens 24-40 23732814-3 2013 Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. Tamoxifen 52-61 heme oxygenase 1 Homo sapiens 104-126 23732814-3 2013 Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. Tamoxifen 52-61 heme oxygenase 1 Homo sapiens 104-108 23732814-3 2013 Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. Tamoxifen 63-66 heme oxygenase 1 Homo sapiens 104-126 23977989-0 2013 Curcumin ameliorates TNF-alpha-induced ICAM-1 expression and subsequent THP-1 adhesiveness via the induction of heme oxygenase-1 in the HaCaT cells. Curcumin 0-8 heme oxygenase 1 Homo sapiens 112-128 23977989-3 2013 Curcumin induced expression of heme oxygenase-1 (HO-1) in the human keratinocyte cell line HaCaT. Curcumin 0-8 heme oxygenase 1 Homo sapiens 31-47 23977989-3 2013 Curcumin induced expression of heme oxygenase-1 (HO-1) in the human keratinocyte cell line HaCaT. Curcumin 0-8 heme oxygenase 1 Homo sapiens 49-53 23977989-5 2013 Curcumin suppressed TNF-alpha- induced ICAM-1 expression and subsequent monocyte adhesion, which were reversed by the addition of tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, or HO-1 knockdown using siRNA. Curcumin 0-8 heme oxygenase 1 Homo sapiens 184-188 23977989-5 2013 Curcumin suppressed TNF-alpha- induced ICAM-1 expression and subsequent monocyte adhesion, which were reversed by the addition of tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, or HO-1 knockdown using siRNA. Curcumin 0-8 heme oxygenase 1 Homo sapiens 193-197 23977989-5 2013 Curcumin suppressed TNF-alpha- induced ICAM-1 expression and subsequent monocyte adhesion, which were reversed by the addition of tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, or HO-1 knockdown using siRNA. tin protoporphyrin IX 130-151 heme oxygenase 1 Homo sapiens 184-188 23977989-5 2013 Curcumin suppressed TNF-alpha- induced ICAM-1 expression and subsequent monocyte adhesion, which were reversed by the addition of tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, or HO-1 knockdown using siRNA. S-Nitroso-N-propionyl-D,L-penicillamine 153-157 heme oxygenase 1 Homo sapiens 184-188 23977989-7 2013 These results suggest that curcumin may exert its anti-inflammatory activity by suppressing the TNF-alpha-induced ICAM-1 expression and subsequent monocyte adhesion via expression of HO-1 in the keratinocytes. Curcumin 27-35 heme oxygenase 1 Homo sapiens 183-187 23671274-2 2013 Here, we report regulation of T-type Ca(2+) channels by carbon monoxide (CO) a HO-1 by-product. Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 79-83 23732814-3 2013 Therefore, we generated mice with cardiac-specific, tamoxifen (TAM)-inducible overexpression of a human HO-1 (hHO-1) transgene (myosin heavy chain (MHC)-HO-1 mice) by breeding mice with cardiac-specific expression of a TAM-inducible Cre recombinase (MHC-Cre mice), with mice containing an hHO-1 transgene preceded by a floxed-stop signal. Tamoxifen 63-66 heme oxygenase 1 Homo sapiens 104-108 24059059-0 2013 [The effect of heme oxygenase-1 on BMSCs damaged by high-concentration glucose]. Glucose 71-78 heme oxygenase 1 Homo sapiens 15-31 24059059-1 2013 This investigation was aimed to explore whether over-expression of 27heme oxygenase-1 (HO-1) could protect bone marrow mesenchymal stem cells(BMSCs)against injury induced by high-concentration glucose. Glucose 193-200 heme oxygenase 1 Homo sapiens 69-85 21809430-5 2013 Our results showed that intracellular ROS were both dose- and time-dependent induced by inorganic arsenic; Cellular Nrf2 protein levels increased rapidly after 2 h of exposure, elevated significantly at 6 h, and reached the maximum at 12 h. The endogenous Nrf2-regulated downstream HO-1 mRNA and protein were also induced dramatically and lasted for as long as 24 h. In addition, intracellular GSH levels elevated in consistent with Nrf2 activation. Reactive Oxygen Species 38-41 heme oxygenase 1 Homo sapiens 282-286 25206471-8 2013 Western blot and real-time PCR analysis showed that the Braintone-containing serum increased the levels of hypoxia-inducible factor 1alpha mRNA and protein, heme oxygenase-1 protein and vascular endothelial growth factor mRNA in oxygen-glucose deprived human umbilical vein endothelial cells. braintone 56-65 heme oxygenase 1 Homo sapiens 157-173 25206471-8 2013 Western blot and real-time PCR analysis showed that the Braintone-containing serum increased the levels of hypoxia-inducible factor 1alpha mRNA and protein, heme oxygenase-1 protein and vascular endothelial growth factor mRNA in oxygen-glucose deprived human umbilical vein endothelial cells. oxygen-glucose 229-243 heme oxygenase 1 Homo sapiens 157-173 23238643-7 2013 We demonstrated that early stage of exposure to IH-induced oxidative and inflammatory stresses leading to acceleration of cell apoptosis via NF-kappaB and Nrf2/HO-1 pathways in endothelial cells, suggesting the potential mechanisms for IH-induced vascular pathogenesis, in resemblance to OSA. Ile-His 48-50 heme oxygenase 1 Homo sapiens 160-164 23238643-7 2013 We demonstrated that early stage of exposure to IH-induced oxidative and inflammatory stresses leading to acceleration of cell apoptosis via NF-kappaB and Nrf2/HO-1 pathways in endothelial cells, suggesting the potential mechanisms for IH-induced vascular pathogenesis, in resemblance to OSA. Ile-His 236-238 heme oxygenase 1 Homo sapiens 160-164 23892599-6 2013 MgNPs-Fe3O4 induced the expression of heme oxygenase-1 at a concentration of 1 mug/mL, and in a dose-dependent manner. mgnps 0-5 heme oxygenase 1 Homo sapiens 38-54 23892599-6 2013 MgNPs-Fe3O4 induced the expression of heme oxygenase-1 at a concentration of 1 mug/mL, and in a dose-dependent manner. ferryl iron 6-11 heme oxygenase 1 Homo sapiens 38-54 23583297-6 2013 In contrast, DIM+SFN or TCDD+SFN induced NQO1 and HMOX1 mRNA expression to levels higher than SFN alone, which was prevented by RNAi-mediated knockdown of AHR. dim+sfn 13-20 heme oxygenase 1 Homo sapiens 50-55 23583297-6 2013 In contrast, DIM+SFN or TCDD+SFN induced NQO1 and HMOX1 mRNA expression to levels higher than SFN alone, which was prevented by RNAi-mediated knockdown of AHR. Polychlorinated Dibenzodioxins 24-28 heme oxygenase 1 Homo sapiens 50-55 23583297-6 2013 In contrast, DIM+SFN or TCDD+SFN induced NQO1 and HMOX1 mRNA expression to levels higher than SFN alone, which was prevented by RNAi-mediated knockdown of AHR. sulforaphane 17-20 heme oxygenase 1 Homo sapiens 50-55 23583297-6 2013 In contrast, DIM+SFN or TCDD+SFN induced NQO1 and HMOX1 mRNA expression to levels higher than SFN alone, which was prevented by RNAi-mediated knockdown of AHR. sulforaphane 29-32 heme oxygenase 1 Homo sapiens 50-55 23583297-7 2013 DIM+SFN but not TCDD+SFN also induced recruitment of ERalpha to NQO1 and HMOX1. dim+sfn 0-7 heme oxygenase 1 Homo sapiens 73-78 23893806-5 2013 Peripheral blood mononuclear cells isolated from asthmatic patients and healthy controls were co-cultured with human bone marrow mesenchymal stem cells which were pretreated with Hemin (the revulsive of Heme Oxygenase-1), Protoporphyrin IX zinc (the inhibitor of Heme Oxygenase-1) and saline. Hemin 179-184 heme oxygenase 1 Homo sapiens 203-219 23893806-5 2013 Peripheral blood mononuclear cells isolated from asthmatic patients and healthy controls were co-cultured with human bone marrow mesenchymal stem cells which were pretreated with Hemin (the revulsive of Heme Oxygenase-1), Protoporphyrin IX zinc (the inhibitor of Heme Oxygenase-1) and saline. Hemin 179-184 heme oxygenase 1 Homo sapiens 263-279 23893806-6 2013 The expression of Heme Oxygenase-1 in MSCs was enhanced by Hemin and inhibited by Protoporphyrin zinc in vitro. Hemin 59-64 heme oxygenase 1 Homo sapiens 18-34 23893806-6 2013 The expression of Heme Oxygenase-1 in MSCs was enhanced by Hemin and inhibited by Protoporphyrin zinc in vitro. zinc protoporphyrin 82-102 heme oxygenase 1 Homo sapiens 18-34 23406266-8 2013 Induction of HO-1 by CoPP (cobalt protoporphyrin IX) reversed these IL-1beta actions. cobaltiprotoporphyrin 21-25 heme oxygenase 1 Homo sapiens 13-17 23406266-8 2013 Induction of HO-1 by CoPP (cobalt protoporphyrin IX) reversed these IL-1beta actions. cobaltiprotoporphyrin 27-51 heme oxygenase 1 Homo sapiens 13-17 21809430-0 2013 Sodium arsenite induced reactive oxygen species generation, nuclear factor (erythroid-2 related) factor 2 activation, heme oxygenase-1 expression, and glutathione elevation in Chang human hepatocytes. sodium arsenite 0-15 heme oxygenase 1 Homo sapiens 118-134 21809430-5 2013 Our results showed that intracellular ROS were both dose- and time-dependent induced by inorganic arsenic; Cellular Nrf2 protein levels increased rapidly after 2 h of exposure, elevated significantly at 6 h, and reached the maximum at 12 h. The endogenous Nrf2-regulated downstream HO-1 mRNA and protein were also induced dramatically and lasted for as long as 24 h. In addition, intracellular GSH levels elevated in consistent with Nrf2 activation. Arsenic 98-105 heme oxygenase 1 Homo sapiens 282-286 24152841-7 2013 Furthermore; iNOS and HO-1 expression and cytokines plasma levels were significantly higher in patients with ACHF as compared to controls group. achf 109-113 heme oxygenase 1 Homo sapiens 22-26 23184650-0 2013 Akt/Nrf2 activated upregulation of heme oxygenase-1 involves in the role of Rg1 against ferrous iron-induced neurotoxicity in SK-N-SH cells. Iron 96-100 heme oxygenase 1 Homo sapiens 35-51 23536331-10 2013 DJ-1 knockout astrocytes showed blunted ability to increase the expression of cellular protective mechanisms against oxidative stress mediated via Nrf-2 and HO-1 in response to exposure to 6-OHDA. Oxidopamine 189-195 heme oxygenase 1 Homo sapiens 157-161 23184650-0 2013 Akt/Nrf2 activated upregulation of heme oxygenase-1 involves in the role of Rg1 against ferrous iron-induced neurotoxicity in SK-N-SH cells. sk-n 126-130 heme oxygenase 1 Homo sapiens 35-51 24024172-5 2013 Interference with the supply of glucose or the pentose phosphate pathway and NADPH generation not only hampers Nrf2-mediated detoxification of reactive oxygen species on the enzyme level but also Nrf2-initiated expression of antioxidant defense proteins, such as glutathione reductase and heme-oxygenase1. Glucose 32-39 heme oxygenase 1 Homo sapiens 289-304 22933385-0 2013 Sanguisorbae radix protects against 6-hydroxydopamine-induced neurotoxicity by regulating NADPH oxidase and NF-E2-related factor-2/heme oxygenase-1 expressions. Oxidopamine 36-53 heme oxygenase 1 Homo sapiens 131-147 22933385-2 2013 The major mechanisms of protection against ROS-induced stress are inhibiting expression of ROS generating genes such as NADPH oxidase (NOX) and increasing expression of endogenous antioxidant genes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 43-46 heme oxygenase 1 Homo sapiens 206-222 22933385-2 2013 The major mechanisms of protection against ROS-induced stress are inhibiting expression of ROS generating genes such as NADPH oxidase (NOX) and increasing expression of endogenous antioxidant genes such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 43-46 heme oxygenase 1 Homo sapiens 224-228 22933385-6 2013 SRE also abolished 6-OHDA-induced ROS by inhibiting NOX expression and by inducing HO-1 expression via NF-E2-related factor-2 activation. Oxidopamine 19-25 heme oxygenase 1 Homo sapiens 83-87 24024172-5 2013 Interference with the supply of glucose or the pentose phosphate pathway and NADPH generation not only hampers Nrf2-mediated detoxification of reactive oxygen species on the enzyme level but also Nrf2-initiated expression of antioxidant defense proteins, such as glutathione reductase and heme-oxygenase1. NADP 77-82 heme oxygenase 1 Homo sapiens 289-304 24024172-5 2013 Interference with the supply of glucose or the pentose phosphate pathway and NADPH generation not only hampers Nrf2-mediated detoxification of reactive oxygen species on the enzyme level but also Nrf2-initiated expression of antioxidant defense proteins, such as glutathione reductase and heme-oxygenase1. Reactive Oxygen Species 143-166 heme oxygenase 1 Homo sapiens 289-304 23840483-4 2013 Since heme molecules are essential for the NADPH oxidase maturation and activity, we therefore investigated the consequences of the modulation of Heme oxygenase-1 (HO-1), the limiting enzyme in heme catabolism, on the IL-1beta signaling pathway and more specifically on Nox4 activity. Heme 6-10 heme oxygenase 1 Homo sapiens 164-168 24024172-5 2013 Interference with the supply of glucose or the pentose phosphate pathway and NADPH generation not only hampers Nrf2-mediated detoxification of reactive oxygen species on the enzyme level but also Nrf2-initiated expression of antioxidant defense proteins, such as glutathione reductase and heme-oxygenase1. Pentosephosphates 47-64 heme oxygenase 1 Homo sapiens 289-304 23840483-8 2013 Therefore, the downregulation of Nox4 activity by HO-1 induction appeared to be mediated by carbon monoxide (CO) generated from the heme degradation process. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 50-54 23840483-8 2013 Therefore, the downregulation of Nox4 activity by HO-1 induction appeared to be mediated by carbon monoxide (CO) generated from the heme degradation process. Carbon Monoxide 109-111 heme oxygenase 1 Homo sapiens 50-54 23840483-8 2013 Therefore, the downregulation of Nox4 activity by HO-1 induction appeared to be mediated by carbon monoxide (CO) generated from the heme degradation process. Heme 132-136 heme oxygenase 1 Homo sapiens 50-54 23604711-5 2013 The induction of HO-1 was preceded by a rise in reactive oxygen species (ROS) and Nrf2 protein expression. Reactive Oxygen Species 48-71 heme oxygenase 1 Homo sapiens 17-21 23604711-5 2013 The induction of HO-1 was preceded by a rise in reactive oxygen species (ROS) and Nrf2 protein expression. Reactive Oxygen Species 73-76 heme oxygenase 1 Homo sapiens 17-21 23604711-7 2013 In addition, the strain-mediated induction of HO-1 and activation of Nrf2 was abolished by the antioxidant N-acetyl-l-cysteine. Acetylcysteine 107-126 heme oxygenase 1 Homo sapiens 46-50 23604711-10 2013 In conclusion, the present study demonstrates that a hemodynamically relevant level of cyclic strain stimulates HO-1 gene expression in ECs via the ROS-Nrf2 signaling pathway to inhibit EC death. Reactive Oxygen Species 148-151 heme oxygenase 1 Homo sapiens 112-116 23665328-6 2013 Treatment of cells with HO-1 inhibitor SnPP IX (tin protoporphyrin IX) attenuated the LPS-mediated responses, HIV-1 replication and secretion of MIP1alpha, MIP1beta, and LD78beta chemokines with little change in surface CCR-5 expression. tin protoporphyrin IX 39-46 heme oxygenase 1 Homo sapiens 24-28 23583009-7 2013 Ethanol-stimulated (100mM) CYP2E1 upregulation was suppressed by quercetin but further enhanced by HO-1 inhibition with resultant heme accumulation. Heme 130-134 heme oxygenase 1 Homo sapiens 99-103 23583009-9 2013 CO donor dose-dependently inactivated CYP2E1 of ethanol-incubated microsome, which was mimicked by HO-1 substrate but abolished by CO scavenger. Ethanol 48-55 heme oxygenase 1 Homo sapiens 99-103 23583009-10 2013 Thus, CYP2E1-mediated ethanol hepatotoxicity was alleviated by quercetin through HO-1 induction. Ethanol 22-29 heme oxygenase 1 Homo sapiens 81-85 23583009-10 2013 Thus, CYP2E1-mediated ethanol hepatotoxicity was alleviated by quercetin through HO-1 induction. Quercetin 63-72 heme oxygenase 1 Homo sapiens 81-85 23583009-2 2013 However, the precise mechanism by which quercetin counteracts CYP2E1-mediated ethanol hepatotoxicity through HO-1 system is still remained unclear. Quercetin 40-49 heme oxygenase 1 Homo sapiens 109-113 23583009-2 2013 However, the precise mechanism by which quercetin counteracts CYP2E1-mediated ethanol hepatotoxicity through HO-1 system is still remained unclear. Ethanol 78-85 heme oxygenase 1 Homo sapiens 109-113 23583009-5 2013 Our data showed that chronic ethanol over-activated CYP2E1 but suppressed HO-1 with concurrent hepatic oxidative damage, which was partially normalized by quercetin (100mg/kg.bw.). Ethanol 29-36 heme oxygenase 1 Homo sapiens 74-78 23583009-5 2013 Our data showed that chronic ethanol over-activated CYP2E1 but suppressed HO-1 with concurrent hepatic oxidative damage, which was partially normalized by quercetin (100mg/kg.bw.). Quercetin 155-164 heme oxygenase 1 Homo sapiens 74-78 23583009-6 2013 Quercetin (100 muM) induced HO-1 and depleted heme pool when incubated to human hepatocytes. Quercetin 0-9 heme oxygenase 1 Homo sapiens 28-32 23583009-7 2013 Ethanol-stimulated (100mM) CYP2E1 upregulation was suppressed by quercetin but further enhanced by HO-1 inhibition with resultant heme accumulation. Ethanol 0-7 heme oxygenase 1 Homo sapiens 99-103 23665328-6 2013 Treatment of cells with HO-1 inhibitor SnPP IX (tin protoporphyrin IX) attenuated the LPS-mediated responses, HIV-1 replication and secretion of MIP1alpha, MIP1beta, and LD78beta chemokines with little change in surface CCR-5 expression. tin protoporphyrin IX 48-69 heme oxygenase 1 Homo sapiens 24-28 23535287-7 2013 Our study indicated that treatment of HepG2 cells with CSA induces Nrf2-dependent ARE activity and gene expression of heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase 1 (NQO1), and glutamate-cysteine ligase modifier subunits by activation of PI3K/AKT, ERK and JNK signaling pathways. 3-hydroxy-4-prenyl-5-methoxystilbene-2-carboxylic acid 55-58 heme oxygenase 1 Homo sapiens 118-134 23525690-9 2013 Taken together, our results suggest that PM(2.5)-induced ROS may function as signaling molecules to activate Nrf2-mediated defenses, such as HO-1 expression, against oxidative stress induced by PM(2.5) through the PI3K/AKT signaling pathway. Reactive Oxygen Species 57-60 heme oxygenase 1 Homo sapiens 141-145 23541843-7 2013 Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, caused a complete restoration of MeHg-mediated inhibition of CYP1A1 activity, induced by TCDD. tin mesoporphyrin 49-66 heme oxygenase 1 Homo sapiens 31-35 23541843-7 2013 Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, caused a complete restoration of MeHg-mediated inhibition of CYP1A1 activity, induced by TCDD. Polychlorinated Dibenzodioxins 157-161 heme oxygenase 1 Homo sapiens 31-35 23541843-8 2013 In addition, transfection of HepG2 cells with siRNA targeting the human HO-1 gene reversed the MeHg-mediated inhibition of TCDD-induced CYP1A1. Polychlorinated Dibenzodioxins 123-127 heme oxygenase 1 Homo sapiens 72-76 23523860-0 2013 Zinc protoporphyrin suppresses cancer cell viability through a heme oxygenase-1-independent mechanism: the involvement of the Wnt/beta-catenin signaling pathway. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 63-79 23523860-1 2013 Zinc protoporphyrin (ZnPP), a known inhibitor of heme oxygenase-1 (HO-1), has been reported to have anticancer activity in both in vitro and in vivo model systems. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 49-65 23412940-6 2013 HO-1 expression was downregulated in cells treated with SB203580 and RAPA. SB 203580 56-64 heme oxygenase 1 Homo sapiens 0-4 23412940-7 2013 HO-1 overexpression inhibited apoptosis and induced G2/M arrest in SB203580 and RAPA-treated cells. SB 203580 67-75 heme oxygenase 1 Homo sapiens 0-4 23412940-8 2013 HO-1 expression was upregulated in the presence of ethanol, and was accompanied by activation of p38MAPK and mTOR. Ethanol 51-58 heme oxygenase 1 Homo sapiens 0-4 23412940-9 2013 However, ethanol-treated cells exposed to HO-1 inhibitor showed no effect on p38MAPK and mTOR activation. Ethanol 9-16 heme oxygenase 1 Homo sapiens 42-46 23523860-1 2013 Zinc protoporphyrin (ZnPP), a known inhibitor of heme oxygenase-1 (HO-1), has been reported to have anticancer activity in both in vitro and in vivo model systems. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 67-71 23523860-1 2013 Zinc protoporphyrin (ZnPP), a known inhibitor of heme oxygenase-1 (HO-1), has been reported to have anticancer activity in both in vitro and in vivo model systems. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 49-65 23523860-1 2013 Zinc protoporphyrin (ZnPP), a known inhibitor of heme oxygenase-1 (HO-1), has been reported to have anticancer activity in both in vitro and in vivo model systems. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 67-71 23523860-2 2013 While the mechanisms of ZnPP"s anticancer activity remain to be elucidated, it is generally believed that ZnPP suppresses tumor growth through inhibition of HO-1 activity. zinc protoporphyrin 106-110 heme oxygenase 1 Homo sapiens 157-161 23523860-3 2013 We examined this hypothesis by altering cellular levels of HO-1 in human ovarian (A2780) and prostate cancer (DU145) cells and found that ZnPP inhibits cancer cell viability through an HO-1-independent mechanism. zinc protoporphyrin 138-142 heme oxygenase 1 Homo sapiens 59-63 23523860-3 2013 We examined this hypothesis by altering cellular levels of HO-1 in human ovarian (A2780) and prostate cancer (DU145) cells and found that ZnPP inhibits cancer cell viability through an HO-1-independent mechanism. zinc protoporphyrin 138-142 heme oxygenase 1 Homo sapiens 185-189 23523860-5 2013 Consistent with these observations, tin protoporphyrin (SnPP), a well-established HO-1 inhibitor, was found to be much less cytotoxic than ZnPP, and docosahexaenoic acid (DHA), an HO-1 inducer, enhanced ZnPP"s cytotoxicity. tin protoporphyrin IX 36-54 heme oxygenase 1 Homo sapiens 82-86 23523860-5 2013 Consistent with these observations, tin protoporphyrin (SnPP), a well-established HO-1 inhibitor, was found to be much less cytotoxic than ZnPP, and docosahexaenoic acid (DHA), an HO-1 inducer, enhanced ZnPP"s cytotoxicity. S-Nitroso-N-propionyl-D,L-penicillamine 56-60 heme oxygenase 1 Homo sapiens 82-86 23523860-5 2013 Consistent with these observations, tin protoporphyrin (SnPP), a well-established HO-1 inhibitor, was found to be much less cytotoxic than ZnPP, and docosahexaenoic acid (DHA), an HO-1 inducer, enhanced ZnPP"s cytotoxicity. S-Nitroso-N-propionyl-D,L-penicillamine 56-60 heme oxygenase 1 Homo sapiens 180-184 23412940-10 2013 The data suggest that ethanol-induced upregulation of HO-1 in oesophageal squamous cell carcinoma is accompanied by the activation of the p38MAPK and mTOR pathways. Ethanol 22-29 heme oxygenase 1 Homo sapiens 54-58 23536693-1 2013 Heme oxygenase-1 (HO-1) is a stress-inducible rate-limiting enzyme in heme degradation that confers cytoprotection against oxidative injury and performs a vital function in the maintenance of cell hemostasis. Heme 70-74 heme oxygenase 1 Homo sapiens 0-16 23536693-1 2013 Heme oxygenase-1 (HO-1) is a stress-inducible rate-limiting enzyme in heme degradation that confers cytoprotection against oxidative injury and performs a vital function in the maintenance of cell hemostasis. Heme 70-74 heme oxygenase 1 Homo sapiens 18-22 23536693-4 2013 Specific inhibitors of mitogen-activated protein kinases (MAPKs), SB203580, PD98059, and SP600125, significantly abolished HO-1 expression. SB 203580 66-74 heme oxygenase 1 Homo sapiens 123-127 23536693-4 2013 Specific inhibitors of mitogen-activated protein kinases (MAPKs), SB203580, PD98059, and SP600125, significantly abolished HO-1 expression. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 76-83 heme oxygenase 1 Homo sapiens 123-127 23536693-4 2013 Specific inhibitors of mitogen-activated protein kinases (MAPKs), SB203580, PD98059, and SP600125, significantly abolished HO-1 expression. pyrazolanthrone 89-97 heme oxygenase 1 Homo sapiens 123-127 23454680-0 2013 Role of alpha-synuclein aggregation and the nuclear factor E2-related factor 2/heme oxygenase-1 pathway in iron-induced neurotoxicity. Iron 107-111 heme oxygenase 1 Homo sapiens 44-95 23454680-6 2013 Consistent with these results, knockdown of alpha-syn expression prevents reduction of Nrf2 and HO-1 by ferrous iron, eliminates alpha-syn aggregates, and protects SK-N-SH cells against ferrous iron-induced cell damage. ferrous iron 104-116 heme oxygenase 1 Homo sapiens 96-100 23649692-0 2013 Reciprocal regulation of cyclooxygenase 2 and heme oxygenase 1 upon arsenic trioxide exposure in normal human lung fibroblast. Arsenic Trioxide 68-84 heme oxygenase 1 Homo sapiens 46-62 23649692-1 2013 Detoxification enzyme heme oxygenase 1 (HO-1) and proinflammation enzyme cyclooxygenase 2 (Cox-2) are key response proteins that function to promote the survival of cells exposed to arsenic trioxide (ATO). Arsenic Trioxide 182-198 heme oxygenase 1 Homo sapiens 22-38 23649692-1 2013 Detoxification enzyme heme oxygenase 1 (HO-1) and proinflammation enzyme cyclooxygenase 2 (Cox-2) are key response proteins that function to promote the survival of cells exposed to arsenic trioxide (ATO). Arsenic Trioxide 182-198 heme oxygenase 1 Homo sapiens 40-44 23649692-1 2013 Detoxification enzyme heme oxygenase 1 (HO-1) and proinflammation enzyme cyclooxygenase 2 (Cox-2) are key response proteins that function to promote the survival of cells exposed to arsenic trioxide (ATO). Arsenic Trioxide 200-203 heme oxygenase 1 Homo sapiens 22-38 23649692-1 2013 Detoxification enzyme heme oxygenase 1 (HO-1) and proinflammation enzyme cyclooxygenase 2 (Cox-2) are key response proteins that function to promote the survival of cells exposed to arsenic trioxide (ATO). Arsenic Trioxide 200-203 heme oxygenase 1 Homo sapiens 40-44 23649692-3 2013 In this study, concomitant upregulation of Cox-2 and HO-1 induced by ATO was observed in normal human lung fibroblasts. Arsenic Trioxide 69-72 heme oxygenase 1 Homo sapiens 53-57 23649692-4 2013 Cox-2 inhibitor NS398 suppressed the upregulation of HO-1, whereas HO-1 inhibitor protoporphyrin IX zinc (II) stimulated the expression of Cox-2. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 16-21 heme oxygenase 1 Homo sapiens 53-57 23649692-4 2013 Cox-2 inhibitor NS398 suppressed the upregulation of HO-1, whereas HO-1 inhibitor protoporphyrin IX zinc (II) stimulated the expression of Cox-2. protoporphyrin IX 82-99 heme oxygenase 1 Homo sapiens 67-71 23649692-6 2013 Our results confirmed the reciprocal regulations between Cox-2 and HO-1 in ATO-treated normal cells and shed light on the understanding of protecting cells from injury caused by ATO while simultaneously decreasing the inflammation responses, which may be related to the carcinogenicity of ATO. Arsenic Trioxide 75-78 heme oxygenase 1 Homo sapiens 67-71 23602216-1 2013 BACKGROUND: The glutathione thymidine repeats [(GT)n] of the heme oxygenase (HO)-1 gene promoter have been shown to be correlated with the incidence of coronary artery disease (CAD), patients with shorter repeats being less likely to have CAD. glutathione thymidine 16-37 heme oxygenase 1 Homo sapiens 61-82 23454680-7 2013 Furthermore, increased HO-1 expression exerts a protective role against ferrous iron. ferrous iron 72-84 heme oxygenase 1 Homo sapiens 23-27 23454680-8 2013 These results support a new hypothesis of synergistic alpha-syn/iron cytotoxicity, whereby ferrous iron induces alpha-syn aggregation and neurotoxicity by inhibiting Nrf2/HO-1. Iron 64-68 heme oxygenase 1 Homo sapiens 171-175 23454680-3 2013 Here, we report that down-regulation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) may contribute to iron-induced alpha-syn aggregation. Iron 128-132 heme oxygenase 1 Homo sapiens 86-102 23454680-8 2013 These results support a new hypothesis of synergistic alpha-syn/iron cytotoxicity, whereby ferrous iron induces alpha-syn aggregation and neurotoxicity by inhibiting Nrf2/HO-1. ferrous iron 91-103 heme oxygenase 1 Homo sapiens 171-175 23454680-9 2013 Inhibition of Nrf2/HO-1 leads to more alpha-syn aggregation and greater toxicity induced by iron, creating a vicious cycle of iron accumulation, alpha-syn aggregation and HO-1 disruption in PD. Iron 92-96 heme oxygenase 1 Homo sapiens 19-23 23454680-3 2013 Here, we report that down-regulation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) may contribute to iron-induced alpha-syn aggregation. Iron 128-132 heme oxygenase 1 Homo sapiens 104-108 23454680-9 2013 Inhibition of Nrf2/HO-1 leads to more alpha-syn aggregation and greater toxicity induced by iron, creating a vicious cycle of iron accumulation, alpha-syn aggregation and HO-1 disruption in PD. Iron 126-130 heme oxygenase 1 Homo sapiens 19-23 23454680-4 2013 In this study, we show that ferrous iron down-regulates Nrf2 and HO-1 in a time-dependent manner in SK-N-SH neuroblastoma cells. ferrous iron 28-40 heme oxygenase 1 Homo sapiens 65-69 23454680-5 2013 Levels of both Nrf2 and HO-1 are decreased even more by ferrous iron in SK-N-SH cells that overexpress alpha-syn and results in greater cell toxicity. ferrous iron 56-68 heme oxygenase 1 Homo sapiens 24-28 23535360-9 2013 Regression analysis revealed that ho-1 and nqo1 expressions were found to be associated with chemical sensitizer reactivity to cysteine, providing evidence of the importance of chemical reactivity, as a part of danger signals, in DC biology. Cysteine 127-135 heme oxygenase 1 Homo sapiens 34-38 23307413-4 2013 Here, we found that multiple factors were released from brain astrocytes (RBA-1) exposed to BK in the conditioned culture media (BK-CM), including ROS, MMP-9, and heme oxygenase-1 (HO-1)/carbon monoxide (CO), leading to neuronal cell (SK-N-SH) death. Carbon Monoxide 187-202 heme oxygenase 1 Homo sapiens 181-185 23307413-4 2013 Here, we found that multiple factors were released from brain astrocytes (RBA-1) exposed to BK in the conditioned culture media (BK-CM), including ROS, MMP-9, and heme oxygenase-1 (HO-1)/carbon monoxide (CO), leading to neuronal cell (SK-N-SH) death. Carbon Monoxide 204-206 heme oxygenase 1 Homo sapiens 181-185 23691207-0 2013 Resveratrol protects C6 astrocyte cell line against hydrogen peroxide-induced oxidative stress through heme oxygenase 1. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 103-119 23691207-7 2013 Resveratrol per se increased heme oxygenase 1 (HO1) expression and extracellular GSH content. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 29-45 23691207-0 2013 Resveratrol protects C6 astrocyte cell line against hydrogen peroxide-induced oxidative stress through heme oxygenase 1. Hydrogen Peroxide 52-69 heme oxygenase 1 Homo sapiens 103-119 23691207-7 2013 Resveratrol per se increased heme oxygenase 1 (HO1) expression and extracellular GSH content. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 47-50 27122714-2 2013 In the present study, we investigated if endothelial expression of cell adhesion molecules (CAMs) is inhibited by fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARalpha) agonist with anti-inflammatory and vascular protective effects, through the regulation of heme oxygenase-1 (HO-1). Fenofibrate 114-125 heme oxygenase 1 Homo sapiens 280-296 23691207-8 2013 In addition, HO1 signaling pathway is involved in the protective effect of resveratrol against H2O2-induced oxidative damage in astroglial cells. Resveratrol 75-86 heme oxygenase 1 Homo sapiens 13-16 23691207-8 2013 In addition, HO1 signaling pathway is involved in the protective effect of resveratrol against H2O2-induced oxidative damage in astroglial cells. Hydrogen Peroxide 95-99 heme oxygenase 1 Homo sapiens 13-16 22689053-7 2013 HO-1 downregulation in renal carcinoma cells induces a mitotic delay at G2/M phase by increasing the intracellular reactive oxygen species and the DNA-damage-induced checkpoint activation. Reactive Oxygen Species 115-138 heme oxygenase 1 Homo sapiens 0-4 23583371-4 2013 Small interfering RNA (siRNA) for HO-1 was transfected into human RPE cell line ARPE-19, and zinc protoporphyrin (ZnPP) was used to inhibit HO-1 activity. zinc protoporphyrin 93-112 heme oxygenase 1 Homo sapiens 140-144 27122714-0 2013 Fenofibrate Modulates HO-1 and Ameliorates Endothelial Expression of Cell Adhesion Molecules in Systolic Heart Failure. Fenofibrate 0-11 heme oxygenase 1 Homo sapiens 22-26 27122714-2 2013 In the present study, we investigated if endothelial expression of cell adhesion molecules (CAMs) is inhibited by fenofibrate, a peroxisome proliferator-activated receptor alpha (PPARalpha) agonist with anti-inflammatory and vascular protective effects, through the regulation of heme oxygenase-1 (HO-1). Fenofibrate 114-125 heme oxygenase 1 Homo sapiens 298-302 27122714-9 2013 Pretreatment with fenofibrate prevented the decrease of HO-1 expression and the activation of NF-kappaB as well as the increase of CAM expression that induced by HF patient serum. Fenofibrate 18-29 heme oxygenase 1 Homo sapiens 56-60 27122714-10 2013 CONCLUSIONS: Our study demonstrated that fenofibrate may exert beneficial effects in patients with systolic HF through regulation of HO-1 expression and amelioration of endothelial activation. Fenofibrate 41-52 heme oxygenase 1 Homo sapiens 133-137 23345623-1 2013 BACKGROUND: The objective of this study was to evaluate the interaction between the length polymorphism of the guanosine thymidine repeat [(GT)n] in the heme oxygenase-1 (HO-1) gene and far-infrared (FIR) therapy on access flow (Qa) and arteriovenous fistula (AVF) patency in hemodialysis (HD) patients. guanosine thymidine 111-130 heme oxygenase 1 Homo sapiens 153-169 23524841-10 2013 CONCLUSIONS: In patients with chronic stable coronary disease, all clinically relevant daily doses of aspirin tested, from 81 to 1300 mg, produce similar and statistically significant increases in HO-1 and decreases in ADMA. Aspirin 102-109 heme oxygenase 1 Homo sapiens 197-201 23288142-0 2013 Inhibition of matrix metalloproteinase-9 expression by docosahexaenoic acid mediated by heme oxygenase 1 in 12-O-tetradecanoylphorbol-13-acetate-induced MCF-7 human breast cancer cells. Docosahexaenoic Acids 55-75 heme oxygenase 1 Homo sapiens 88-104 23288142-0 2013 Inhibition of matrix metalloproteinase-9 expression by docosahexaenoic acid mediated by heme oxygenase 1 in 12-O-tetradecanoylphorbol-13-acetate-induced MCF-7 human breast cancer cells. Tetradecanoylphorbol Acetate 108-144 heme oxygenase 1 Homo sapiens 88-104 23261939-6 2013 The heme oxygenase-1 (HO-1) induction in response to H2O2 and MPP(+) treatment was impaired by the overexpression of the PINK1 P209A mutant. Hydrogen Peroxide 53-57 heme oxygenase 1 Homo sapiens 4-20 23261939-6 2013 The heme oxygenase-1 (HO-1) induction in response to H2O2 and MPP(+) treatment was impaired by the overexpression of the PINK1 P209A mutant. Hydrogen Peroxide 53-57 heme oxygenase 1 Homo sapiens 22-26 23345623-1 2013 BACKGROUND: The objective of this study was to evaluate the interaction between the length polymorphism of the guanosine thymidine repeat [(GT)n] in the heme oxygenase-1 (HO-1) gene and far-infrared (FIR) therapy on access flow (Qa) and arteriovenous fistula (AVF) patency in hemodialysis (HD) patients. guanosine thymidine 111-130 heme oxygenase 1 Homo sapiens 171-175 23419834-10 2013 Treatment with hemin, a HO-1 inducer, and with [Ru(CO)3Cl2]2, a CO donor, decreased LPS-stimulated iNOS induction and NO production. Hemin 15-20 heme oxygenase 1 Homo sapiens 24-28 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Thymine 107-114 heme oxygenase 1 Homo sapiens 144-160 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Thymine 107-114 heme oxygenase 1 Homo sapiens 167-171 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Thymine 107-114 heme oxygenase 1 Homo sapiens 273-277 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Guanine 115-122 heme oxygenase 1 Homo sapiens 144-160 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Guanine 115-122 heme oxygenase 1 Homo sapiens 167-171 23571756-1 2013 Using a luciferase reporter assay, we previously demonstrated that a Z-DNA-forming sequence of alternating thymine-guanine repeats in the human heme oxygenase-1 gene (HO-1) promoter is involved in nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2)-mediated HO-1 promoter activation. Guanine 115-122 heme oxygenase 1 Homo sapiens 273-277 23571756-5 2013 Using this detection system, we demonstrated that the glutathione-depleting agent, diethyl maleate, induced Nrf2-dependent Z-DNA formation in the HO-1 promoter, but not in the thioredoxin reductase 1 gene promoter. Glutathione 54-65 heme oxygenase 1 Homo sapiens 146-150 23571756-5 2013 Using this detection system, we demonstrated that the glutathione-depleting agent, diethyl maleate, induced Nrf2-dependent Z-DNA formation in the HO-1 promoter, but not in the thioredoxin reductase 1 gene promoter. diethyl maleate 83-98 heme oxygenase 1 Homo sapiens 146-150 23403272-0 2013 Paeoniflorin protects human EA.hy926 endothelial cells against gamma-radiation induced oxidative injury by activating the NF-E2-related factor 2/heme oxygenase-1 pathway. peoniflorin 0-12 heme oxygenase 1 Homo sapiens 145-161 23602847-8 2013 However, the protective effects of LXA4 were completely blocked by transfection of the cells with HO-1 siRNA, and were partially but significantly blocked by pretreatment of the cells with various blockers of K(+) channels. lipoxin A4 35-39 heme oxygenase 1 Homo sapiens 98-102 23602847-9 2013 The LXA4-induced expressions of HO-1 in the cells were also inhibited by HO-1 siRNA and various blockers of K(+) channels. lipoxin A4 4-8 heme oxygenase 1 Homo sapiens 32-36 23602847-9 2013 The LXA4-induced expressions of HO-1 in the cells were also inhibited by HO-1 siRNA and various blockers of K(+) channels. lipoxin A4 4-8 heme oxygenase 1 Homo sapiens 73-77 23602847-10 2013 The inhibitory effects of LXA4 on enhanced TNF-alpha levels induced by H/R injury were abolished by transfection of the cells with HO-1 siRNA. lipoxin A4 26-30 heme oxygenase 1 Homo sapiens 131-135 23535229-6 2013 The autophagy inhibitor 3-methyladenine significantly inhibited HO-1 up-regulation and increased the rate of cell death in cells treated with P-VO2, while the HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) enhanced the occurrence of cell death in the P-VO2-treated cells while having no effect on the autophagic response induced by P-VO2. 3-methyladenine 24-39 heme oxygenase 1 Homo sapiens 64-68 23328493-8 2013 Finally, DSC inhibited H2O2-induced changes of Bcl-2, Bax, and caspase-3 expression, and all of these effects were reversed by HO-1 silencing. Hydrogen Peroxide 23-27 heme oxygenase 1 Homo sapiens 127-131 23535229-6 2013 The autophagy inhibitor 3-methyladenine significantly inhibited HO-1 up-regulation and increased the rate of cell death in cells treated with P-VO2, while the HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) enhanced the occurrence of cell death in the P-VO2-treated cells while having no effect on the autophagic response induced by P-VO2. protoporphyrin IX 174-191 heme oxygenase 1 Homo sapiens 159-163 23535229-6 2013 The autophagy inhibitor 3-methyladenine significantly inhibited HO-1 up-regulation and increased the rate of cell death in cells treated with P-VO2, while the HO-1 inhibitor protoporphyrin IX zinc (II) (ZnPP) enhanced the occurrence of cell death in the P-VO2-treated cells while having no effect on the autophagic response induced by P-VO2. zinc protoporphyrin 203-207 heme oxygenase 1 Homo sapiens 159-163 23570271-5 2013 Genetic studies have provided an opportunity to identify the proteins that link vitamin D to ALS pathology, including major histocompatibility complex (MHC) class II molecules, toll-like receptors, poly(ADP-ribose) polymerase-1, heme oxygenase-1, and calcium-binding proteins, as well as the reduced form of nicotinamide adenine dinucleotide phosphate. Vitamin D 80-89 heme oxygenase 1 Homo sapiens 229-245 23328493-6 2013 In addition, DSC concentration-dependently induced HO-1 expression associated with nuclear translocation of nuclear factor-erythroid 2 related factor 2 (Nrf-2), while the effect of DSC was inhibited by a phosphoinositide 3-kinase (PI3K) inhibitor LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 247-255 heme oxygenase 1 Homo sapiens 51-55 23290930-0 2013 Hydroquinone regulates hemeoxygenase-1 expression via modulation of Src kinase activity through thiolation of cysteine residues. hydroquinone 0-12 heme oxygenase 1 Homo sapiens 23-38 23328493-7 2013 Furthermore, the protective effect of DSC on H2O2-induced cell death was abolished by HO-1 inhibitor ZnPP, but was mimicked by carbon monoxide-releasing moiety CORM-3 or HO-1 by-product bilirubin. Hydrogen Peroxide 45-49 heme oxygenase 1 Homo sapiens 86-90 23090785-1 2013 Heme oxygenase-1 (HO-1) is both beneficial and detrimental to the host in some viral infections by catalyzing the conversion of heme to biliverdin, iron, and carbon monoxide. Heme 128-132 heme oxygenase 1 Homo sapiens 18-22 23090785-1 2013 Heme oxygenase-1 (HO-1) is both beneficial and detrimental to the host in some viral infections by catalyzing the conversion of heme to biliverdin, iron, and carbon monoxide. Biliverdine 136-146 heme oxygenase 1 Homo sapiens 18-22 23090785-1 2013 Heme oxygenase-1 (HO-1) is both beneficial and detrimental to the host in some viral infections by catalyzing the conversion of heme to biliverdin, iron, and carbon monoxide. Iron 148-152 heme oxygenase 1 Homo sapiens 18-22 23291594-10 2013 C18 acid 1 induced the expression of ARE-regulated cytoprotective genes, including NAD(P)H:quinone oxidoreductase 1, heme oxygenase 1, thioredoxin reductase 1, both subunits of the glutamate-cysteine ligase (catalytic subunit and modifier subunit), and the cystine/glutamate exchange transporter, in IMR-32 human neuroblastoma cells. c18 acid 1 0-10 heme oxygenase 1 Homo sapiens 117-133 23146110-12 2013 Andrographolide up-regulated ARE-regulated gene targets including glutamate-cysteine ligase catalytic (GCLC) subunit, GCL modifier (GCLM) subunit, GPx, GR and heme oxygenase-1 in BEAS-2B cells in response to CSE. andrographolide 0-15 heme oxygenase 1 Homo sapiens 159-175 23090785-1 2013 Heme oxygenase-1 (HO-1) is both beneficial and detrimental to the host in some viral infections by catalyzing the conversion of heme to biliverdin, iron, and carbon monoxide. Carbon Monoxide 158-173 heme oxygenase 1 Homo sapiens 18-22 23090785-6 2013 HO-1 overexpression in BGC-823 cells caused the cells containing Blasticidin-resistant gene driven by SV40 promoter to grow slowly under Blasticidin screening, compared with control groups. blasticidin S 65-76 heme oxygenase 1 Homo sapiens 0-4 23090785-6 2013 HO-1 overexpression in BGC-823 cells caused the cells containing Blasticidin-resistant gene driven by SV40 promoter to grow slowly under Blasticidin screening, compared with control groups. blasticidin S 137-148 heme oxygenase 1 Homo sapiens 0-4 23290930-0 2013 Hydroquinone regulates hemeoxygenase-1 expression via modulation of Src kinase activity through thiolation of cysteine residues. Cysteine 110-118 heme oxygenase 1 Homo sapiens 23-38 23350672-4 2013 Heme-hemopexin is relatively resistant to damage by ROS and retains its ability to induce the cytoprotective HO1 after exposure to tert-butylhydroperoxide, although induction is impaired, but not eliminated, by exposure to high concentrations of H(2)O(2) in vitro. tert-Butylhydroperoxide 131-154 heme oxygenase 1 Homo sapiens 109-112 23274124-3 2013 Thiamine also affects HIV through non-genomic factors, i.e., matrix metalloproteinase, vascular endothelial growth factor, heme oxygenase 1, the prostaglandins, cyclooxygenase 2, reactive oxygen species, and nitric oxide. Thiamine 0-8 heme oxygenase 1 Homo sapiens 123-139 23567851-0 2013 Homocysteine induces heme oxygenase-1 expression via transcription factor Nrf2 activation in HepG2 cell. Homocysteine 0-12 heme oxygenase 1 Homo sapiens 21-37 23567851-8 2013 Real time RT-PCR and Western-blotting were performed to evaluate whether homocysteine was able to induce mRNA and protein expression of HO-1. Homocysteine 73-85 heme oxygenase 1 Homo sapiens 136-140 23567851-11 2013 (iii) Real time RT-PCR and Western-blotting revealed increased mRNA and protein expression of inducible gene HO-1 after treatment with homocysteine. Homocysteine 135-147 heme oxygenase 1 Homo sapiens 109-113 23567851-13 2013 Therefore, induction of Nrf2-ARE-dependent expression of HO-1 could be a therapeutic option for hepatic cells damage induced by homocysteine. Homocysteine 128-140 heme oxygenase 1 Homo sapiens 57-61 23350672-4 2013 Heme-hemopexin is relatively resistant to damage by ROS and retains its ability to induce the cytoprotective HO1 after exposure to tert-butylhydroperoxide, although induction is impaired, but not eliminated, by exposure to high concentrations of H(2)O(2) in vitro. Hydrogen Peroxide 246-254 heme oxygenase 1 Homo sapiens 109-112 22946793-10 2013 Moreover, the protective role of melatonin against damage to endothelial tight junction integrity was addressed by ZO-1 expression, paralleled with the restored heme oxygenase-1 levels during OGD. Melatonin 33-42 heme oxygenase 1 Homo sapiens 161-177 23350672-8 2013 Hemopexin sequesters heme, thus preventing unregulated heme uptake that leads to toxicity; it safely delivers heme to neuronal cells; and it activates the induction of proteins including HO1 and hAPP that keep heme and iron at safe levels in neurons. Iron 219-223 heme oxygenase 1 Homo sapiens 187-190 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Heme 59-63 heme oxygenase 1 Homo sapiens 77-81 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Carbon Monoxide 134-149 heme oxygenase 1 Homo sapiens 59-75 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Carbon Monoxide 134-149 heme oxygenase 1 Homo sapiens 77-81 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Carbon Monoxide 151-153 heme oxygenase 1 Homo sapiens 59-75 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Carbon Monoxide 151-153 heme oxygenase 1 Homo sapiens 77-81 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Biliverdine 156-166 heme oxygenase 1 Homo sapiens 59-75 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Biliverdine 156-166 heme oxygenase 1 Homo sapiens 77-81 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Iron 176-180 heme oxygenase 1 Homo sapiens 59-75 23403148-4 2013 One of the key molecules implicated in sPE pathogenesis is heme oxygenase-1 (HO-1), a rate-limiting enzyme that breaks down heme into carbon monoxide (CO), biliverdin and free iron. Iron 176-180 heme oxygenase 1 Homo sapiens 77-81 23403148-5 2013 CO and bilirubin (a downstream product of biliverdin processing) account for the angiogenic, vasodilatory and anti-oxidant properties of HO-1. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 137-141 23403148-5 2013 CO and bilirubin (a downstream product of biliverdin processing) account for the angiogenic, vasodilatory and anti-oxidant properties of HO-1. Bilirubin 7-16 heme oxygenase 1 Homo sapiens 137-141 23403148-5 2013 CO and bilirubin (a downstream product of biliverdin processing) account for the angiogenic, vasodilatory and anti-oxidant properties of HO-1. Biliverdine 42-52 heme oxygenase 1 Homo sapiens 137-141 23403148-6 2013 These collective actions of the heme breakdown metabolites generated by HO-1 offer protection against cytotoxicity, inflammation, hypoxia and other forms of cellular stress that are central to the pathogenesis of sPE. Heme 32-36 heme oxygenase 1 Homo sapiens 72-76 23128353-6 2013 Glucosamine at 50 mM was demonstrated to elevate both the mRNA and protein expression of p53 and heme oxygenase-1 (HO-1), but also caused a reduction in p21 protein expression. Glucosamine 0-11 heme oxygenase 1 Homo sapiens 97-113 23538684-4 2013 RESULTS: Changes in mRNA levels were most numerous and striking at 6 h after heme treatment but were similar and still numerous at 24 h. After 6 h of heme exposure, the increase in heme oxygenase 1 gene expression was 60-fold by mRNA and 88-fold by quantitative reverse transcription-polymerase chain reaction. Heme 77-81 heme oxygenase 1 Homo sapiens 181-197 23128353-6 2013 Glucosamine at 50 mM was demonstrated to elevate both the mRNA and protein expression of p53 and heme oxygenase-1 (HO-1), but also caused a reduction in p21 protein expression. Glucosamine 0-11 heme oxygenase 1 Homo sapiens 115-119 23128353-8 2013 Altogether, our results suggest that a high dose of glucosamine may inhibit cell proliferation through apoptosis and disturb cell cycle progression with a halt at G(0)/G(1) phase, and that this occurs, at least in part, by a reduction in Rb phosphorylation together with modulation of p21, p53 and HO-1 expression, and nuclear p21 accumulation. Glucosamine 52-63 heme oxygenase 1 Homo sapiens 298-302 23419114-0 2013 Dihydroquercetin (DHQ) induced HO-1 and NQO1 expression against oxidative stress through the Nrf2-dependent antioxidant pathway. taxifolin 0-16 heme oxygenase 1 Homo sapiens 31-35 23419114-0 2013 Dihydroquercetin (DHQ) induced HO-1 and NQO1 expression against oxidative stress through the Nrf2-dependent antioxidant pathway. taxifolin 18-21 heme oxygenase 1 Homo sapiens 31-35 23419114-7 2013 DHQ upregulated the Nrf2-related antioxidant genes heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), and glutamate-cysteine ligase modifier subunits. taxifolin 0-3 heme oxygenase 1 Homo sapiens 51-67 23419114-7 2013 DHQ upregulated the Nrf2-related antioxidant genes heme oxygenase-1 (HO-1), NAD(P)H quinone oxidoreductase-1 (NQO1), and glutamate-cysteine ligase modifier subunits. taxifolin 0-3 heme oxygenase 1 Homo sapiens 69-73 23324179-9 2013 Metformin (the best known clinical activator of AMPK) suppressed EMT induction through inhibition of ROS via induction of heme oxygenase-1 and endogenous antioxidant thioredoxin. Metformin 0-9 heme oxygenase 1 Homo sapiens 122-138 23324179-9 2013 Metformin (the best known clinical activator of AMPK) suppressed EMT induction through inhibition of ROS via induction of heme oxygenase-1 and endogenous antioxidant thioredoxin. Reactive Oxygen Species 101-104 heme oxygenase 1 Homo sapiens 122-138 23538684-4 2013 RESULTS: Changes in mRNA levels were most numerous and striking at 6 h after heme treatment but were similar and still numerous at 24 h. After 6 h of heme exposure, the increase in heme oxygenase 1 gene expression was 60-fold by mRNA and 88-fold by quantitative reverse transcription-polymerase chain reaction. Heme 150-154 heme oxygenase 1 Homo sapiens 181-197 23090292-1 2013 The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). cho 195-198 heme oxygenase 1 Homo sapiens 89-105 23322908-4 2013 Vitamin D also exerts its effects on AD by regulating calcium-sensing receptor expression, enhancing amyloid-beta peptides clearance, interleukin 10, downregulating matrix metalloproteinases, upregulating heme oxygenase 1, and suppressing the reduced form of nicotinamide adenine dinucleotide phosphate expression. Vitamin D 0-9 heme oxygenase 1 Homo sapiens 205-221 23090292-1 2013 The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). Carbohydrates 180-193 heme oxygenase 1 Homo sapiens 89-105 23090292-1 2013 The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). Carbohydrates 180-193 heme oxygenase 1 Homo sapiens 107-112 23090292-1 2013 The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). cho 195-198 heme oxygenase 1 Homo sapiens 107-112 23090292-1 2013 The aim of this study was to observe the intracellular heat shock protein 72 (HSP72) and heme oxygenase-1 (HSP32) response to prolonged interval cycling following the ingestion of carbohydrates (CHO) and sodium bicarbonate (NaHCO(3)). Sodium Bicarbonate 204-222 heme oxygenase 1 Homo sapiens 89-105 23090292-5 2013 Exogenous CHO had no influence on either HSP72 or HSP32, but the ingestion of NaHCO(3) significantly attenuated HSP32 in monocytes and lymphocytes (p <= 0.042). Sodium Bicarbonate 78-86 heme oxygenase 1 Homo sapiens 112-117 22717332-7 2013 The HO-1 activator, CoppIX, reversed iNOS/NO up-regulation and HO-1 down-regulation induced by IFN-alpha. coppix 20-26 heme oxygenase 1 Homo sapiens 4-8 22865541-8 2013 Here, we demonstrate that celastrol, but not classical heat shock treatment, is effective in inducing a set of neuroprotective Hsps in cultures derived from cerebral cortices, including Hsp70, Hsp27 and Hsp32. celastrol 26-35 heme oxygenase 1 Homo sapiens 203-208 22717332-7 2013 The HO-1 activator, CoppIX, reversed iNOS/NO up-regulation and HO-1 down-regulation induced by IFN-alpha. coppix 20-26 heme oxygenase 1 Homo sapiens 63-67 23525626-9 2013 These inhibitory effects BL were almost completely abolished by CC and partly by tin protoporphyrin-IX, a competitive inhibitor of HO-1. tin protoporphyrin IX 81-102 heme oxygenase 1 Homo sapiens 131-135 22849812-0 2013 Anti-inflammatory effect of pachymic acid promotes odontoblastic differentiation via HO-1 in dental pulp cells. pachymic acid 28-41 heme oxygenase 1 Homo sapiens 85-89 23281030-10 2013 CONCLUSION: We showed that attenuation of Nrf2 and HO-1 expression through induction of miR-132 and miR-200c by OTA elevates ROS levels and profibrotic TGFbeta expression. Reactive Oxygen Species 125-128 heme oxygenase 1 Homo sapiens 51-55 22264017-9 2013 HMOX1 may thus be a useful biomarker of TOPO-QDOT QD exposure across cell types and species. topo-qdot 40-49 heme oxygenase 1 Homo sapiens 0-5 23318726-0 2013 Butein protects human dental pulp cells from hydrogen peroxide-induced oxidative toxicity via Nrf2 pathway-dependent heme oxygenase-1 expressions. butein 0-6 heme oxygenase 1 Homo sapiens 117-133 23318726-0 2013 Butein protects human dental pulp cells from hydrogen peroxide-induced oxidative toxicity via Nrf2 pathway-dependent heme oxygenase-1 expressions. Hydrogen Peroxide 45-62 heme oxygenase 1 Homo sapiens 117-133 23318726-8 2013 In addition, butein-dependent HO-1 expression was required for the inhibition of H2O2-induced cell death and ROS generation. Hydrogen Peroxide 81-85 heme oxygenase 1 Homo sapiens 30-34 23318726-8 2013 In addition, butein-dependent HO-1 expression was required for the inhibition of H2O2-induced cell death and ROS generation. Reactive Oxygen Species 109-112 heme oxygenase 1 Homo sapiens 30-34 23318726-10 2013 Treatment of HDP cells with a c-Jun NH2-terminal kinase (JNK) inhibitor also reduced butein-induced HO-1 expression, and butein treatment led to increased JNK phosphorylation. butein 85-91 heme oxygenase 1 Homo sapiens 100-104 23525626-0 2013 beta-Lapachone, a substrate of NAD(P)H:quinone oxidoreductase, induces anti-inflammatory heme oxygenase-1 via AMP-activated protein kinase activation in RAW264.7 macrophages. beta-lapachone 0-14 heme oxygenase 1 Homo sapiens 89-105 23525626-6 2013 5-Aminoimidazole-4-carboxamide-1-beta-D-ribofuranoside, an AMPK activator, also induced HO-1 expression. acadesine 0-54 heme oxygenase 1 Homo sapiens 88-92 23281030-0 2013 Cross-talk between microRNAs, nuclear factor E2-related factor 2, and heme oxygenase-1 in ochratoxin A-induced toxic effects in renal proximal tubular epithelial cells. ochratoxin A 90-102 heme oxygenase 1 Homo sapiens 70-86 23281030-2 2013 We investigated the cross-talk between microRNAs, nuclear factor E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in ochratoxin A-mediated effects. ochratoxin A 124-136 heme oxygenase 1 Homo sapiens 97-113 23281030-2 2013 We investigated the cross-talk between microRNAs, nuclear factor E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) in ochratoxin A-mediated effects. ochratoxin A 124-136 heme oxygenase 1 Homo sapiens 115-119 23498787-9 2013 Increased HIF-1alpha activation and HO-1 expression were observed in fenoldopam pretreatment group. Fenoldopam 69-79 heme oxygenase 1 Homo sapiens 36-40 23498787-11 2013 CONCLUSION: Donor preconditioning by fenoldopam exerts renoprotection in grafts, at least in part, through HIF-1alpha activation and HO-1 expression. Fenoldopam 37-47 heme oxygenase 1 Homo sapiens 133-137 22849812-2 2013 In this study, pachymic acid from mushroom Formitopsis niagra is examined to determine whether it affects pulpal inflammation and promotes odontogenesis via HO-1 gene expression. pachymic acid 15-28 heme oxygenase 1 Homo sapiens 157-161 22849812-5 2013 To understand the mechanism of pachymic acid via HO-1 induction, the cells were treated with zinc protoporphyrin IX (ZnPP: HO-1 inhibitor). pachymic acid 31-44 heme oxygenase 1 Homo sapiens 49-53 22849812-5 2013 To understand the mechanism of pachymic acid via HO-1 induction, the cells were treated with zinc protoporphyrin IX (ZnPP: HO-1 inhibitor). zinc protoporphyrin 93-115 heme oxygenase 1 Homo sapiens 123-127 22849812-7 2013 However, the HDPCs treated with pachymic acid affected anti-inflammatory effect and induction of odontoblast differentiation through increasing HO-1 expression. pachymic acid 32-45 heme oxygenase 1 Homo sapiens 144-148 22849812-11 2013 CONCLUSIONS: The pachymic acid showed anti-inflammatory function and odontoblast differentiation via HO-1 pathway. pachymic acid 17-30 heme oxygenase 1 Homo sapiens 101-105 23333393-3 2013 In addition, berberine also induced heme oxygenase (HO)-1 in a dose-dependent manner, which was mediated through activation of p38 MAPK and NF-E2-related factor 2 (Nrf2) signaling cascade in macrophages. Berberine 13-22 heme oxygenase 1 Homo sapiens 36-57 23068042-0 2013 Development of a heme sensor using fluorescently labeled heme oxygenase-1. Heme 17-21 heme oxygenase 1 Homo sapiens 57-73 23025298-7 2013 RECENT ADVANCES: Among the latter, the heme oxygenase-1 (HO-1) pathway is likely to contribute to the established and powerful antioxidant/anti-inflammatory properties of polyphenols. Polyphenols 171-182 heme oxygenase 1 Homo sapiens 39-55 23025298-7 2013 RECENT ADVANCES: Among the latter, the heme oxygenase-1 (HO-1) pathway is likely to contribute to the established and powerful antioxidant/anti-inflammatory properties of polyphenols. Polyphenols 171-182 heme oxygenase 1 Homo sapiens 57-61 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 15-19 heme oxygenase 1 Homo sapiens 98-114 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 15-19 heme oxygenase 1 Homo sapiens 116-120 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 98-114 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 116-120 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 98-114 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 116-120 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 98-114 23068042-2 2013 To detect free heme at low concentrations, we developed a heme sensor using fluorescently labeled heme oxygenase-1 (HO-1), an enzyme that catalyzes oxidative heme degradation and has a high affinity for heme. Heme 58-62 heme oxygenase 1 Homo sapiens 116-120 23068042-4 2013 Each of the three fluorescently labeled HO-1s exhibits a 1:1 binding stoichiometry and an absorption spectrum similar to that of the heme complex of the wild-type HO-1. Heme 133-137 heme oxygenase 1 Homo sapiens 40-44 23068042-5 2013 Titration of the labeled proteins with hemin resulted in fluorescence quenching in a hemin concentration-dependent manner, presumably due to an energy transfer from the fluorophore to the heme bound to HO-1. Hemin 39-44 heme oxygenase 1 Homo sapiens 202-206 23068042-5 2013 Titration of the labeled proteins with hemin resulted in fluorescence quenching in a hemin concentration-dependent manner, presumably due to an energy transfer from the fluorophore to the heme bound to HO-1. Hemin 85-90 heme oxygenase 1 Homo sapiens 202-206 23068042-5 2013 Titration of the labeled proteins with hemin resulted in fluorescence quenching in a hemin concentration-dependent manner, presumably due to an energy transfer from the fluorophore to the heme bound to HO-1. Heme 188-192 heme oxygenase 1 Homo sapiens 202-206 22704780-9 2013 Western blot analysis revealed that catechin stimulated a time-dependent increase in both the nuclear factor erythroid 2-related factor 2 and total heme oxygenase-1 protein expression in Int-407 cells. Catechin 36-44 heme oxygenase 1 Homo sapiens 148-164 23392716-6 2013 In vitro treatment with CoPPIX significantly increased the expression of HO-1 in iPECs in a dose-dependent manner. cobaltiprotoporphyrin 24-30 heme oxygenase 1 Homo sapiens 73-77 23392716-7 2013 Over-expression of HO-1 was successfully achieved by incubation of iPECs with either 50 or 80 mumol/L of CoPPIX. cobaltiprotoporphyrin 105-111 heme oxygenase 1 Homo sapiens 19-23 23255485-6 2013 In Caco-2 cells, treatment with genistein markedly attenuated H(2)O(2) -induced peroxide formation; this amelioration was reversed by buthionine sulfoximine(GCLC inhibitor) and zinc protoporphyrin(HO-1 inhibitor). Genistein 32-41 heme oxygenase 1 Homo sapiens 197-201 23255485-6 2013 In Caco-2 cells, treatment with genistein markedly attenuated H(2)O(2) -induced peroxide formation; this amelioration was reversed by buthionine sulfoximine(GCLC inhibitor) and zinc protoporphyrin(HO-1 inhibitor). Hydrogen Peroxide 62-70 heme oxygenase 1 Homo sapiens 197-201 23255485-6 2013 In Caco-2 cells, treatment with genistein markedly attenuated H(2)O(2) -induced peroxide formation; this amelioration was reversed by buthionine sulfoximine(GCLC inhibitor) and zinc protoporphyrin(HO-1 inhibitor). zinc protoporphyrin 177-196 heme oxygenase 1 Homo sapiens 197-201 23329300-0 2013 Berberine protects 6-hydroxydopamine-induced human dopaminergic neuronal cell death through the induction of heme oxygenase-1. Berberine 0-9 heme oxygenase 1 Homo sapiens 109-125 23255485-11 2013 These results suggest the involvement of ERK1/2, PKC, and Nrf2 in inducing HO-1 and GCLC by genistein. Genistein 92-101 heme oxygenase 1 Homo sapiens 75-79 23329300-0 2013 Berberine protects 6-hydroxydopamine-induced human dopaminergic neuronal cell death through the induction of heme oxygenase-1. Oxidopamine 19-36 heme oxygenase 1 Homo sapiens 109-125 23329300-4 2013 BBR also upregulated heme oxygenase-1 (HO-1) expression, which conferred protection against 6-OHDA-induced dopaminergic neuron injury and besides, effect of BBR on HO-1 was reversed by siRNA-Nrf2. Oxidopamine 92-98 heme oxygenase 1 Homo sapiens 21-37 23255485-12 2013 CONCLUSION: Our studies show that genistein up-regulated HO-1 and GCLC expression through the EKR1/2 and PKC /Nrf2 pathways during oxidative stress. Genistein 34-43 heme oxygenase 1 Homo sapiens 57-61 23026155-2 2013 The intracellular levels of CO can increase under stressful conditions following the induction of HO-1 (heme oxygnase-1), a ubiquitous enzyme responsible for the catabolism of heme. Heme 104-108 heme oxygenase 1 Homo sapiens 98-102 23085367-6 2013 Xanthohumol, beside the induction of GSTs and HO-1, significantly elevated NQO1 expression in concert with p53 level in normal hepatocytes. xanthohumol 0-11 heme oxygenase 1 Homo sapiens 46-50 23143154-13 2013 Quercetin also reduced the CSE- and H(2)O(2)-induced upregulation of ROS and HO-1 protein in differentiated OFs and preadipocyte OFs. Quercetin 0-9 heme oxygenase 1 Homo sapiens 77-81 22716961-9 2013 Moreover, the use of Nrf2 small interfering RNA transfection and HO-1- or PPARgamma-specific antagonists (Znpp and GW9662, respectively) blocked the protective effects of genistein on endothelial cell viability during oxidative stress. Genistein 171-180 heme oxygenase 1 Homo sapiens 65-70 23143154-13 2013 Quercetin also reduced the CSE- and H(2)O(2)-induced upregulation of ROS and HO-1 protein in differentiated OFs and preadipocyte OFs. Hydrogen Peroxide 36-44 heme oxygenase 1 Homo sapiens 77-81 23295066-4 2013 Ultimately, physiological studies unveiled the endogenous production of CO, particularly by the heme oxygenase (HO)-1 enzyme, recognizing CO as a beneficial gas when used at therapeutic doses. Carbon Monoxide 72-74 heme oxygenase 1 Homo sapiens 96-117 23097500-0 2013 Structural insights into human heme oxygenase-1 inhibition by potent and selective azole-based compounds. Azoles 83-88 heme oxygenase 1 Homo sapiens 31-47 23221079-8 2013 In contrast, HO-1 protein was upregulated by treatment with both hydrogen peroxide and light. Hydrogen Peroxide 65-82 heme oxygenase 1 Homo sapiens 13-17 23131562-0 2013 Omega-3 polyunsaturated fatty acid has an anti-oxidant effect via the Nrf-2/HO-1 pathway in 3T3-L1 adipocytes. omega-3 polyunsaturated fatty acid 0-34 heme oxygenase 1 Homo sapiens 76-80 23131562-6 2013 Knockdown of nuclear factor erythroid 2-related factor 2 (Nrf-2) significantly reduced EPA, DHA or 4-HHE-induced HO-1 mRNA and protein expression. Eicosapentaenoic Acid 87-90 heme oxygenase 1 Homo sapiens 113-117 23131562-6 2013 Knockdown of nuclear factor erythroid 2-related factor 2 (Nrf-2) significantly reduced EPA, DHA or 4-HHE-induced HO-1 mRNA and protein expression. Docosahexaenoic Acids 92-95 heme oxygenase 1 Homo sapiens 113-117 23131562-7 2013 Also, pretreatment with omega3-PUFA prevented H(2)O(2)-induced cytotoxicity in a HO-1 dependent manner. Hydrogen Peroxide 46-54 heme oxygenase 1 Homo sapiens 81-85 23131562-8 2013 In conclusion, treatment with EPA and DHA induced HO-1 through the activation of Nrf-2 and prevented oxidative stress in 3T3-L1 adipocytes. Eicosapentaenoic Acid 30-33 heme oxygenase 1 Homo sapiens 50-54 23131562-8 2013 In conclusion, treatment with EPA and DHA induced HO-1 through the activation of Nrf-2 and prevented oxidative stress in 3T3-L1 adipocytes. Docosahexaenoic Acids 38-41 heme oxygenase 1 Homo sapiens 50-54 22989377-3 2013 We investigated HO-1-overexpressing A549 cells and find that, as expected, HO-1-overexpressing cells are resistant to killing by hydrogen peroxide. Hydrogen Peroxide 129-146 heme oxygenase 1 Homo sapiens 75-79 22989377-5 2013 However, HO-1-overexpressing cells contain only ~25% as much "loose" (probably redox active) iron. Iron 93-97 heme oxygenase 1 Homo sapiens 9-13 22989377-6 2013 Indeed, inhibition of ferritin synthesis [via siRNA (small interfering RNA) directed at the ferritin heavy chain] sensitizes the HO-1-overexpressing cells to peroxide killing. Peroxides 158-166 heme oxygenase 1 Homo sapiens 129-133 22989377-9 2013 We conclude that, at least in many cases, the cytoprotective effects of HO-1 induction or forced overexpression may derive from elevated expression of ferritin and consequent reduction of redox active "loose" iron. Iron 209-213 heme oxygenase 1 Homo sapiens 72-76 23046979-0 2013 Diallyl tetrasulfane activates both the eIF2alpha and Nrf2/HO-1 pathways. diallyl tetrasulfane 0-20 heme oxygenase 1 Homo sapiens 59-63 23046979-8 2013 DATTS treatment of HCT116 cells also caused an up-regulation of phospho-eIF2alpha, nuclear Nrf2 and HO-1 protein levels in a time and concentration-dependent manner. datts 0-5 heme oxygenase 1 Homo sapiens 100-104 23046979-11 2013 CONCLUSIONS: DATTS activates the ROS-eIF2alpha/Nrf2 HO-1 signaling cascades leading to the up-regulation of HO-1. datts 13-18 heme oxygenase 1 Homo sapiens 52-56 23046979-11 2013 CONCLUSIONS: DATTS activates the ROS-eIF2alpha/Nrf2 HO-1 signaling cascades leading to the up-regulation of HO-1. datts 13-18 heme oxygenase 1 Homo sapiens 108-112 23046979-11 2013 CONCLUSIONS: DATTS activates the ROS-eIF2alpha/Nrf2 HO-1 signaling cascades leading to the up-regulation of HO-1. Reactive Oxygen Species 33-36 heme oxygenase 1 Homo sapiens 52-56 23046979-11 2013 CONCLUSIONS: DATTS activates the ROS-eIF2alpha/Nrf2 HO-1 signaling cascades leading to the up-regulation of HO-1. Reactive Oxygen Species 33-36 heme oxygenase 1 Homo sapiens 108-112 23811562-6 2013 Other assays revealed that, in keratinocytes, sauchinone decreased reactive oxygen species (ROS) production and increased glutathione levels and heme oxygenase-1. sauchinone 46-56 heme oxygenase 1 Homo sapiens 145-161 23152113-8 2013 THG213.29 increased mRNA expression of heme-oxygenase 1, Bcl2, and FGF-2 in renal cortex; correspondingly, in EP(4)-transfected HEK293 cells, THG213.29 augmented FGF-2 and abrogated EP(4)-dependent overexpression of inflammatory IL-6 and of apoptotic death domain-associated protein and BCL2-associated agonist of cell death. thg213 0-6 heme oxygenase 1 Homo sapiens 39-55 23546295-7 2013 The antioxidant potential of EA might be directly correlated with the increased expression of HO-1 and NQO1, whose expression may have surmounted the oxidative stress generated by PQ. Paraquat 180-182 heme oxygenase 1 Homo sapiens 94-98 23291378-0 2013 Genistein inhibits ox-LDL-induced VCAM-1, ICAM-1 and MCP-1 expression of HUVECs through heme oxygenase-1. Genistein 0-9 heme oxygenase 1 Homo sapiens 88-104 23291378-7 2013 RESULTS: Pretreatment with genistein markedly reduced ox-LDL-induced MCP-1, VCAM-1 and ICAM-1 secretion and mRNA transcription, which was further decreased by the inducer of HO and reversed by the inhibitor of HO; additionally, the effects were accompanied with upregulating HO-1 mRNA and protein expression and markedly abolished with Nrf2 siRNA. Genistein 27-36 heme oxygenase 1 Homo sapiens 275-279 24191253-0 2013 Antidiabetic potential of the heme oxygenase-1 inducer curcumin analogues. Curcumin 55-63 heme oxygenase 1 Homo sapiens 30-46 24009858-7 2013 Interestingly, we found that treatment of finasteride induced the expression of Nrf2 and HO-1 proteins in PC-3 cells. Finasteride 42-53 heme oxygenase 1 Homo sapiens 89-93 23738323-6 2013 Furthermore, mollugin induced the activation of p38, ERK, and JNK and the expression of heme oxygenase-1 (HO-1) and nuclear factor E2-related factor 2 (Nrf2). rubimaillin 13-21 heme oxygenase 1 Homo sapiens 88-104 23738323-6 2013 Furthermore, mollugin induced the activation of p38, ERK, and JNK and the expression of heme oxygenase-1 (HO-1) and nuclear factor E2-related factor 2 (Nrf2). rubimaillin 13-21 heme oxygenase 1 Homo sapiens 106-110 23738323-7 2013 Mollugin-induced growth inhibition and apoptosis of HO-1 were reversed by an HO-1 inhibitor and Nrf2 siRNA. rubimaillin 0-8 heme oxygenase 1 Homo sapiens 52-56 23738323-7 2013 Mollugin-induced growth inhibition and apoptosis of HO-1 were reversed by an HO-1 inhibitor and Nrf2 siRNA. rubimaillin 0-8 heme oxygenase 1 Homo sapiens 77-81 23738323-8 2013 Collectively, this is the first report to demonstrate the effectiveness of mollugin as a candidate for a chemotherapeutic agent in OSCCs via the upregulation of the HO-1 and Nrf2 pathways and the downregulation of NF- kappa B. rubimaillin 75-83 heme oxygenase 1 Homo sapiens 165-169 24009858-0 2013 Finasteride Increases the Expression of Hemoxygenase-1 (HO-1) and NF-E2-Related Factor-2 (Nrf2) Proteins in PC-3 Cells: Implication of Finasteride-Mediated High-Grade Prostate Tumor Occurrence. Finasteride 0-11 heme oxygenase 1 Homo sapiens 40-54 23868099-6 2013 Additionally, we found that the zinc-induced HO-1 gene transcription can be enhanced by clioquinol, a zinc ionophore, and reversed by pretreatment with TPEN, a known zinc chelator, indicating that an increase in intracellular zinc levels is responsible for this induction. Clioquinol 88-98 heme oxygenase 1 Homo sapiens 45-49 24009858-0 2013 Finasteride Increases the Expression of Hemoxygenase-1 (HO-1) and NF-E2-Related Factor-2 (Nrf2) Proteins in PC-3 Cells: Implication of Finasteride-Mediated High-Grade Prostate Tumor Occurrence. Finasteride 0-11 heme oxygenase 1 Homo sapiens 56-60 23087099-6 2013 Such upregulated HO-1 levels were effective in conferring protection against H(2)O(2)-induced cell death and in promoting the proangiogenic phenotype of HMEC-1 cells. Hydrogen Peroxide 77-85 heme oxygenase 1 Homo sapiens 17-21 23868099-6 2013 Additionally, we found that the zinc-induced HO-1 gene transcription can be enhanced by clioquinol, a zinc ionophore, and reversed by pretreatment with TPEN, a known zinc chelator, indicating that an increase in intracellular zinc levels is responsible for this induction. N,N,N',N'-tetrakis(2-pyridylmethyl)ethylenediamine 152-156 heme oxygenase 1 Homo sapiens 45-49 23739547-0 2013 Tertiary-butylhydroquinone upregulates expression of ATP-binding cassette transporter A1 via nuclear factor E2-related factor 2/heme oxygenase-1 signaling in THP-1 macrophage-derived foam cells. 2-Butylhydroquinone 9-26 heme oxygenase 1 Homo sapiens 108-144 24247298-0 2013 Clofibrate induces heme oxygenase 1 expression through a PPARalpha-independent mechanism in human cancer cells. Clofibrate 0-10 heme oxygenase 1 Homo sapiens 19-35 24247298-2 2013 This study examined the effect of clofibrate on heme oxygenase-1 (HO-1) gene expression in A2780 (human ovarian cancer) and DU145 (human prostate cancer) cells. Clofibrate 34-44 heme oxygenase 1 Homo sapiens 48-64 24247298-2 2013 This study examined the effect of clofibrate on heme oxygenase-1 (HO-1) gene expression in A2780 (human ovarian cancer) and DU145 (human prostate cancer) cells. Clofibrate 34-44 heme oxygenase 1 Homo sapiens 66-70 24247298-3 2013 METHODS AND RESULTS: We demonstrate that clofibrate induces HO-1 expression in a concentration- and time-dependent manner. Clofibrate 41-51 heme oxygenase 1 Homo sapiens 60-64 24247298-4 2013 The induction of HO-1 by clofibrate was detected at both mRNA and protein levels and the HO-1 gene promoter activity was also dramatically induced by clofibrate, indicating that clofibrate up-regulates HO-1 gene transcription. Clofibrate 25-35 heme oxygenase 1 Homo sapiens 17-21 24247298-4 2013 The induction of HO-1 by clofibrate was detected at both mRNA and protein levels and the HO-1 gene promoter activity was also dramatically induced by clofibrate, indicating that clofibrate up-regulates HO-1 gene transcription. Clofibrate 150-160 heme oxygenase 1 Homo sapiens 89-93 24247298-4 2013 The induction of HO-1 by clofibrate was detected at both mRNA and protein levels and the HO-1 gene promoter activity was also dramatically induced by clofibrate, indicating that clofibrate up-regulates HO-1 gene transcription. Clofibrate 150-160 heme oxygenase 1 Homo sapiens 89-93 24247298-4 2013 The induction of HO-1 by clofibrate was detected at both mRNA and protein levels and the HO-1 gene promoter activity was also dramatically induced by clofibrate, indicating that clofibrate up-regulates HO-1 gene transcription. Clofibrate 150-160 heme oxygenase 1 Homo sapiens 89-93 24247298-4 2013 The induction of HO-1 by clofibrate was detected at both mRNA and protein levels and the HO-1 gene promoter activity was also dramatically induced by clofibrate, indicating that clofibrate up-regulates HO-1 gene transcription. Clofibrate 150-160 heme oxygenase 1 Homo sapiens 89-93 24247298-5 2013 Surprisingly, the induction of HO-1 by clofibrate was mediated by the Nrf2 signaling pathway, not by the PPARalpha pathway. Clofibrate 39-49 heme oxygenase 1 Homo sapiens 31-35 24247298-6 2013 This was primarily demonstrated by siRNA knockdown of Nrf2 expression that significantly attenuated clofibrate-induced HO-1 gene transcription, and siRNA knockdown of PPARalpha that had no effect on clofibrate-induced HO-1 promoter activity. Clofibrate 100-110 heme oxygenase 1 Homo sapiens 119-123 24247298-7 2013 Furthermore, deletion of the antioxidant response elements (AREs) in the HO-1 gene promoter diminished clofibrate-induced HO-1 transcription and deletion of the PPAR response elements (PPREs) had no such effect. Clofibrate 103-113 heme oxygenase 1 Homo sapiens 73-77 24247298-7 2013 Furthermore, deletion of the antioxidant response elements (AREs) in the HO-1 gene promoter diminished clofibrate-induced HO-1 transcription and deletion of the PPAR response elements (PPREs) had no such effect. Clofibrate 103-113 heme oxygenase 1 Homo sapiens 122-126 24247298-9 2013 CONCLUSION: Clofibrate induces HO-1 gene expression in cancer cells through a PPARalpha-independent mechanism and the Nrf2 signaling pathway is indispensible for this induction. Clofibrate 12-22 heme oxygenase 1 Homo sapiens 31-35 23739547-5 2013 Furthermore, tBHQ reduced calpain-mediated ABCA1 proteolysis via activation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). 2-tert-butylhydroquinone 13-17 heme oxygenase 1 Homo sapiens 125-141 23739547-5 2013 Furthermore, tBHQ reduced calpain-mediated ABCA1 proteolysis via activation of nuclear factor E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1). 2-tert-butylhydroquinone 13-17 heme oxygenase 1 Homo sapiens 143-147 23739547-6 2013 Inhibition of HO-1 with a pharmacological inhibitor or siRNA and knockdown of Nrf2 suppressed the stimulatory effects of tBHQ on ABCA1 expression and calpain activity. 2-tert-butylhydroquinone 121-125 heme oxygenase 1 Homo sapiens 14-18 23739547-7 2013 CONCLUSIONS: Nrf2/HO-1 signaling is required for the regulation by tBHQ of ABCA1 expression and cholesterol efflux in macrophage-derived foam cells and an antiatherogenic role of tBHQ is suggested. 2-tert-butylhydroquinone 67-71 heme oxygenase 1 Homo sapiens 18-22 23739547-7 2013 CONCLUSIONS: Nrf2/HO-1 signaling is required for the regulation by tBHQ of ABCA1 expression and cholesterol efflux in macrophage-derived foam cells and an antiatherogenic role of tBHQ is suggested. Cholesterol 96-107 heme oxygenase 1 Homo sapiens 18-22 23739547-7 2013 CONCLUSIONS: Nrf2/HO-1 signaling is required for the regulation by tBHQ of ABCA1 expression and cholesterol efflux in macrophage-derived foam cells and an antiatherogenic role of tBHQ is suggested. 2-tert-butylhydroquinone 179-183 heme oxygenase 1 Homo sapiens 18-22 23720291-6 2013 The rate-controlling step of heme breakdown is catalyzed by heme oxygenase (HMOX), of which there are two isoforms, called HMOX1 and HMOX2. Heme 29-33 heme oxygenase 1 Homo sapiens 123-128 24327821-4 2013 But the potential role of resveratrol against As2O3 in heart via nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) is unclear. Resveratrol 26-37 heme oxygenase 1 Homo sapiens 120-136 23092324-5 2013 In the present study we investigated the combined effect of L-Carnosine and EGCG on the activation of two stress-responsive pathways: HO-1 and Hsp72 (the inducible form of Hsp70), which play an important role in cytoprotection against oxidative stress-induced cell damage. epigallocatechin gallate 76-80 heme oxygenase 1 Homo sapiens 134-138 23092324-6 2013 We demonstrated that the neuroprotective effects of EGCG and L-Carnosine are achieved through the modulation of HO-1/Hsp72 systems. epigallocatechin gallate 52-56 heme oxygenase 1 Homo sapiens 112-116 23092325-0 2013 Nuclear translocation of heme oxygenase-1 confers resistance to imatinib in chronic myeloid leukemia cells. Imatinib Mesylate 64-72 heme oxygenase 1 Homo sapiens 25-41 23092325-5 2013 The aim of the present study was to investigate the effect of HO-1 expression on cell proliferation and apoptosis in chronic myeloid leukemia cells, K562 and LAMA-84 cell lines following imatinib treatment. Imatinib Mesylate 187-195 heme oxygenase 1 Homo sapiens 62-66 23092325-6 2013 Cells were incubated for 24h with Imatinib (1 muM) alone or in combination with Hemin (10muM), an inducer of HO-1. Imatinib Mesylate 34-42 heme oxygenase 1 Homo sapiens 109-113 23092325-6 2013 Cells were incubated for 24h with Imatinib (1 muM) alone or in combination with Hemin (10muM), an inducer of HO-1. Hemin 80-85 heme oxygenase 1 Homo sapiens 109-113 23092325-7 2013 In addition, cells were also treated with HO byproducts, bilirubin and carbon monoxide (CO), or with a protease inhibitor (Ed64) to inhibit HO-1 nuclear translocation. ed64 123-127 heme oxygenase 1 Homo sapiens 140-144 23092325-8 2013 Pharmacological induction of HO-1 was able to overcome the effect of imatinib. Imatinib Mesylate 69-77 heme oxygenase 1 Homo sapiens 29-33 23092325-12 2013 Finally, imatinib was able to increase the formation of cellular reactive oxygen species (ROS) and this effect was reversed by HO-1 induction or the addition of N-acetylcisteine (NAC). Imatinib Mesylate 9-17 heme oxygenase 1 Homo sapiens 127-131 23092325-12 2013 Finally, imatinib was able to increase the formation of cellular reactive oxygen species (ROS) and this effect was reversed by HO-1 induction or the addition of N-acetylcisteine (NAC). Reactive Oxygen Species 65-88 heme oxygenase 1 Homo sapiens 127-131 23092325-12 2013 Finally, imatinib was able to increase the formation of cellular reactive oxygen species (ROS) and this effect was reversed by HO-1 induction or the addition of N-acetylcisteine (NAC). Reactive Oxygen Species 90-93 heme oxygenase 1 Homo sapiens 127-131 23092325-13 2013 In conclusion, the protective effect of HO-1 on imatinib-induced cytotoxicity does not involve its enzymatic byproducts, but rather the nuclear translocation of HO-1 following proteolytic cleavage. Imatinib Mesylate 48-56 heme oxygenase 1 Homo sapiens 40-44 23092325-13 2013 In conclusion, the protective effect of HO-1 on imatinib-induced cytotoxicity does not involve its enzymatic byproducts, but rather the nuclear translocation of HO-1 following proteolytic cleavage. Imatinib Mesylate 48-56 heme oxygenase 1 Homo sapiens 161-165 23092328-0 2013 Heme Oxygenase-1 and breast cancer resistance protein protect against heme-induced toxicity. Heme 70-74 heme oxygenase 1 Homo sapiens 0-16 23092328-5 2013 We postulated that overexpression of Heme Oxygenase-1 (HO-1) and Breast Cancer Resistance Protein (BCRP) would protect against heme-induced cytotoxicity. Heme 127-131 heme oxygenase 1 Homo sapiens 37-53 23092328-5 2013 We postulated that overexpression of Heme Oxygenase-1 (HO-1) and Breast Cancer Resistance Protein (BCRP) would protect against heme-induced cytotoxicity. Heme 127-131 heme oxygenase 1 Homo sapiens 55-59 23092328-6 2013 HO-1 is a heme-degrading enzyme generating carbon monoxide, iron, and biliverdin/bilirubin, while BCRP is a heme efflux transporter. Carbon Monoxide 43-58 heme oxygenase 1 Homo sapiens 0-4 23092328-6 2013 HO-1 is a heme-degrading enzyme generating carbon monoxide, iron, and biliverdin/bilirubin, while BCRP is a heme efflux transporter. Iron 60-64 heme oxygenase 1 Homo sapiens 0-4 23746277-9 2013 Historically, the first molecules used as non selective HO-1 inhibitors were metalloporphyrins (Mps). Metalloporphyrins 77-94 heme oxygenase 1 Homo sapiens 56-60 23746277-9 2013 Historically, the first molecules used as non selective HO-1 inhibitors were metalloporphyrins (Mps). Metalloporphyrins 96-99 heme oxygenase 1 Homo sapiens 56-60 23746277-10 2013 The subsequent development of the imidazole-dioxolane derivatives afforded the first generation of non-porphyrin based, isozyme selective HO-1 inhibitors. imidazole-dioxolane 34-53 heme oxygenase 1 Homo sapiens 138-142 23746277-10 2013 The subsequent development of the imidazole-dioxolane derivatives afforded the first generation of non-porphyrin based, isozyme selective HO-1 inhibitors. Porphyrins 103-112 heme oxygenase 1 Homo sapiens 138-142 23092328-6 2013 HO-1 is a heme-degrading enzyme generating carbon monoxide, iron, and biliverdin/bilirubin, while BCRP is a heme efflux transporter. Biliverdine 70-80 heme oxygenase 1 Homo sapiens 0-4 23092328-6 2013 HO-1 is a heme-degrading enzyme generating carbon monoxide, iron, and biliverdin/bilirubin, while BCRP is a heme efflux transporter. Bilirubin 81-90 heme oxygenase 1 Homo sapiens 0-4 23092328-9 2013 Heme-induced cell death was significantly attenuated when cells overexpressed HO-1, BCRP, or both. Heme 0-4 heme oxygenase 1 Homo sapiens 78-82 23092328-11 2013 Also cells treated with the anti-oxidants N-acetylcysteine or HO-effector molecule bilirubin showed protection against heme insults, which may explain the increased protection by HO-1 compared to BCRP. Acetylcysteine 42-58 heme oxygenase 1 Homo sapiens 179-183 23092328-11 2013 Also cells treated with the anti-oxidants N-acetylcysteine or HO-effector molecule bilirubin showed protection against heme insults, which may explain the increased protection by HO-1 compared to BCRP. Bilirubin 83-92 heme oxygenase 1 Homo sapiens 179-183 23092328-11 2013 Also cells treated with the anti-oxidants N-acetylcysteine or HO-effector molecule bilirubin showed protection against heme insults, which may explain the increased protection by HO-1 compared to BCRP. Heme 119-123 heme oxygenase 1 Homo sapiens 179-183 23092328-12 2013 In conclusion, both HO-1 and BCRP protect against heme-induced toxicity and may thus form novel therapeutic targets for heme-mediated pathologies. Heme 50-54 heme oxygenase 1 Homo sapiens 20-24 23092328-12 2013 In conclusion, both HO-1 and BCRP protect against heme-induced toxicity and may thus form novel therapeutic targets for heme-mediated pathologies. Heme 120-124 heme oxygenase 1 Homo sapiens 20-24 24302971-0 2013 Acetylshikonin, a Novel AChE Inhibitor, Inhibits Apoptosis via Upregulation of Heme Oxygenase-1 Expression in SH-SY5Y Cells. acetylshikonin 0-14 heme oxygenase 1 Homo sapiens 79-95 24302971-7 2013 Furthermore, we identified for the first time that the upregulation of heme oxygenase 1 by acetylshikonin is a key step mediating its antiapoptotic activity from oxidative stress in SH-SY5Y cells. acetylshikonin 91-105 heme oxygenase 1 Homo sapiens 71-87 24327821-4 2013 But the potential role of resveratrol against As2O3 in heart via nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) is unclear. Resveratrol 26-37 heme oxygenase 1 Homo sapiens 138-142 24327821-8 2013 Furthermore, resveratrol significantly prohibited the downregulation of both Nrf2 and HO-1 gene expressions that were downregulated by As2O3, whereas resveratrol did not alter As2O3-induced nitric oxide formation. Resveratrol 13-24 heme oxygenase 1 Homo sapiens 86-90 24327821-8 2013 Furthermore, resveratrol significantly prohibited the downregulation of both Nrf2 and HO-1 gene expressions that were downregulated by As2O3, whereas resveratrol did not alter As2O3-induced nitric oxide formation. Arsenic Trioxide 135-140 heme oxygenase 1 Homo sapiens 86-90 24327821-9 2013 Thus, the protective role of resveratrol against As2O3-induced cardiotoxicity is implemented by the maintenance of redox homeostasis (Nrf2-HO-1 pathway) and facilitating arsenic efflux. Resveratrol 29-40 heme oxygenase 1 Homo sapiens 139-143 24327821-9 2013 Thus, the protective role of resveratrol against As2O3-induced cardiotoxicity is implemented by the maintenance of redox homeostasis (Nrf2-HO-1 pathway) and facilitating arsenic efflux. Arsenic Trioxide 49-54 heme oxygenase 1 Homo sapiens 139-143 23573137-0 2013 The protective effect of alpha-lipoic Acid in lipopolysaccharide-induced acute lung injury is mediated by heme oxygenase-1. Thioctic Acid 25-42 heme oxygenase 1 Homo sapiens 106-122 23573137-6 2013 Interestingly, treatment with ALA significantly increased nuclear factor-erythroid 2-related factor 2 (Nrf2) activation and HO-1 expression in lungs of ALI. Thioctic Acid 30-33 heme oxygenase 1 Homo sapiens 124-128 23573137-3 2013 The aim of study was to test the hypothesis that the protection of ALA against lipopolysaccharide-(LPS-) induced acute lung injury (ALI) is mediated by HO-1. Thioctic Acid 67-70 heme oxygenase 1 Homo sapiens 152-156 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. tin protoporphyrin IX 35-56 heme oxygenase 1 Homo sapiens 18-22 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. tin protoporphyrin IX 35-56 heme oxygenase 1 Homo sapiens 68-72 23063670-1 2013 The aim of this study was to evaluate the effect of phloretamide (PA), an apple constituent, on the activation of the Nrf2 transcription factor and the expression of its target genes: glutathione S-transferases (GSTs), NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) in normal human THLE-2 hepatocytes and the hepatoma HepG2 cell line. phloretamide 52-64 heme oxygenase 1 Homo sapiens 281-285 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. S-Nitroso-N-propionyl-D,L-penicillamine 58-62 heme oxygenase 1 Homo sapiens 18-22 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. S-Nitroso-N-propionyl-D,L-penicillamine 58-62 heme oxygenase 1 Homo sapiens 68-72 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. Thioctic Acid 131-134 heme oxygenase 1 Homo sapiens 18-22 23573137-7 2013 However, blocking HO-1 activity by tin protoporphyrin IX (SnPP), an HO-1 inhibitor, markedly abolished these beneficial effects of ALA in LPS-induced ALI. Thioctic Acid 131-134 heme oxygenase 1 Homo sapiens 68-72 23573137-8 2013 These results suggest that the protection mechanism of ALA may be through HO-1 induction and in turn suppressing NF- kappa B-mediated inflammatory responses. Thioctic Acid 55-58 heme oxygenase 1 Homo sapiens 74-78 23063670-1 2013 The aim of this study was to evaluate the effect of phloretamide (PA), an apple constituent, on the activation of the Nrf2 transcription factor and the expression of its target genes: glutathione S-transferases (GSTs), NAD(P)H:quinone oxidoreductase-1 (NQO1) and heme oxygenase-1 (HO-1) in normal human THLE-2 hepatocytes and the hepatoma HepG2 cell line. phloretamide 66-68 heme oxygenase 1 Homo sapiens 281-285 24094245-3 2013 Electrophoretic mobility-shift assays (EMSAs) showed that ARE was a prominent element for HO-1 induction after low-concentration HBCDs exposure. hexabromocyclododecane 129-134 heme oxygenase 1 Homo sapiens 90-94 24094245-4 2013 The relationship between PI3K/Akt pathway and Nrf2/HO-1 axis was demonstrated by the finding that pretreatment with PI3K inhibitors (wortmannin, LY294002) attenuated the upregulation of Nrf2 expression induced by HBCDs exposure. Wortmannin 133-143 heme oxygenase 1 Homo sapiens 51-55 24485305-1 2013 Haem oxygenase 1 (HO-1) plays a pivotal role in metabolic stress protecting cells in dependence on reactive oxygen species. Reactive Oxygen Species 99-122 heme oxygenase 1 Homo sapiens 0-22 24094245-4 2013 The relationship between PI3K/Akt pathway and Nrf2/HO-1 axis was demonstrated by the finding that pretreatment with PI3K inhibitors (wortmannin, LY294002) attenuated the upregulation of Nrf2 expression induced by HBCDs exposure. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 145-153 heme oxygenase 1 Homo sapiens 51-55 24094245-4 2013 The relationship between PI3K/Akt pathway and Nrf2/HO-1 axis was demonstrated by the finding that pretreatment with PI3K inhibitors (wortmannin, LY294002) attenuated the upregulation of Nrf2 expression induced by HBCDs exposure. hexabromocyclododecane 213-218 heme oxygenase 1 Homo sapiens 51-55 23103292-1 2013 Heme oxygenase (HO)-1, the inducible isoform of the first and rate-limiting enzyme of heme degradation, affords anti-inflammatory protection via its cell-type-specific effects in endothelial cells (ECs). Heme 86-90 heme oxygenase 1 Homo sapiens 0-21 23103292-7 2013 Reactive oxygen species, which were generated by NADPH oxidase and in turn activated the phosphatidylinositol 3-kinase (PI3K)/Akt cascade, were involved in this upregulation of HO-1 gene expression. Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 177-181 23476651-0 2013 Zinc Protoporphyrin Upregulates Heme Oxygenase-1 in PC-3 Cells via the Stress Response Pathway. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 32-48 23476651-1 2013 Zinc protoporphyrin IX (ZnPP), a naturally occurring molecule formed in iron deficiency or lead poisoning, is a potent competitive inhibitor of heme oxygenase-1 (HO-1). zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 144-160 23476651-1 2013 Zinc protoporphyrin IX (ZnPP), a naturally occurring molecule formed in iron deficiency or lead poisoning, is a potent competitive inhibitor of heme oxygenase-1 (HO-1). zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 162-166 23476651-1 2013 Zinc protoporphyrin IX (ZnPP), a naturally occurring molecule formed in iron deficiency or lead poisoning, is a potent competitive inhibitor of heme oxygenase-1 (HO-1). zinc protoporphyrin 24-28 heme oxygenase 1 Homo sapiens 144-160 23476651-1 2013 Zinc protoporphyrin IX (ZnPP), a naturally occurring molecule formed in iron deficiency or lead poisoning, is a potent competitive inhibitor of heme oxygenase-1 (HO-1). zinc protoporphyrin 24-28 heme oxygenase 1 Homo sapiens 162-166 23476651-3 2013 However, the effect of ZnPP on HO-1 expression is controversial. zinc protoporphyrin 23-27 heme oxygenase 1 Homo sapiens 31-35 23476651-5 2013 The objective of this study is to investigate the effect of ZnPP on HO-1 expression in prostate cancer PC-3 cells. zinc protoporphyrin 60-64 heme oxygenase 1 Homo sapiens 68-72 23476651-6 2013 Incubation of PC-3 cells with 10 mu M ZnPP for 4 h showed only a slight induction of HO-1 mRNA and protein, but the induction was high after 16 h and was maintained through 48 h of incubation. zinc protoporphyrin 39-43 heme oxygenase 1 Homo sapiens 86-90 23476651-8 2013 Of the various protein kinase inhibitors and antioxidant tested, only Ro 31-8220 abrogated ZnPP-induced HO-1 expression, suggesting that activation of HO-1 gene by ZnPP may involve protein kinase C (PKC). zinc protoporphyrin 91-95 heme oxygenase 1 Homo sapiens 104-108 23476651-8 2013 Of the various protein kinase inhibitors and antioxidant tested, only Ro 31-8220 abrogated ZnPP-induced HO-1 expression, suggesting that activation of HO-1 gene by ZnPP may involve protein kinase C (PKC). zinc protoporphyrin 91-95 heme oxygenase 1 Homo sapiens 151-155 23476651-8 2013 Of the various protein kinase inhibitors and antioxidant tested, only Ro 31-8220 abrogated ZnPP-induced HO-1 expression, suggesting that activation of HO-1 gene by ZnPP may involve protein kinase C (PKC). zinc protoporphyrin 164-168 heme oxygenase 1 Homo sapiens 104-108 23476651-8 2013 Of the various protein kinase inhibitors and antioxidant tested, only Ro 31-8220 abrogated ZnPP-induced HO-1 expression, suggesting that activation of HO-1 gene by ZnPP may involve protein kinase C (PKC). zinc protoporphyrin 164-168 heme oxygenase 1 Homo sapiens 151-155 24148794-0 2013 Heme oxygenase-1 mediates the anti-inflammatory effect of molecular hydrogen in LPS-stimulated RAW 264.7 macrophages. Hydrogen 68-76 heme oxygenase 1 Homo sapiens 0-16 23569422-0 2013 Propofol-induced protection of SH-SY5Y cells against hydrogen peroxide is associated with the HO-1 via the ERK pathway. Propofol 0-8 heme oxygenase 1 Homo sapiens 94-98 23569422-0 2013 Propofol-induced protection of SH-SY5Y cells against hydrogen peroxide is associated with the HO-1 via the ERK pathway. Hydrogen Peroxide 53-70 heme oxygenase 1 Homo sapiens 94-98 23569422-5 2013 Additionally, the potential roles of ERK, p 38 MAPK and JNK in the regulation of propofol-induced endogenous HO-1 expression in SH-SY5Y cells were estimated by Western blotting assays. Propofol 81-89 heme oxygenase 1 Homo sapiens 109-113 24148794-10 2013 Furthermore, H2 treatment could also dose-dependently increase the HO-1 protein expression and activity at 3 h, 6 h, 12 h and 24 h in LPS-activated macrophages. Hydrogen 13-15 heme oxygenase 1 Homo sapiens 67-71 24148794-11 2013 In addition, blockade of HO-1 activity with ZnPP-IX partly reversed the anti-inflammatory effect of H2 in LPS-stimulated macrophages. Hydrogen 100-102 heme oxygenase 1 Homo sapiens 25-29 24148794-2 2013 In the present study, we investigated whether heme oxygenase-1 (HO-1) contributes to the anti-inflammatory effect of H2 in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Hydrogen 117-119 heme oxygenase 1 Homo sapiens 46-62 24148794-12 2013 CONCLUSIONS: Molecular hydrogen exerts a regulating role in the release of pro- and anti-inflammatory cytokines in LPS-stimulated macrophages, and this effect is at least partly mediated by HO-1 expression and activation. Hydrogen 23-31 heme oxygenase 1 Homo sapiens 190-194 24148794-2 2013 In the present study, we investigated whether heme oxygenase-1 (HO-1) contributes to the anti-inflammatory effect of H2 in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Hydrogen 117-119 heme oxygenase 1 Homo sapiens 64-68 22901690-0 2013 Docosahexaenoic acid inhibition of inflammation is partially via cross-talk between Nrf2/heme oxygenase 1 and IKK/NF-kappaB pathways. Docosahexaenoic Acids 0-20 heme oxygenase 1 Homo sapiens 89-105 22945906-0 2013 Peroxisome proliferator-activated receptor delta agonist attenuates nicotine suppression effect on human mesenchymal stem cell-derived osteogenesis and involves increased expression of heme oxygenase-1. Nicotine 68-76 heme oxygenase 1 Homo sapiens 185-201 22819551-3 2013 Here, we report that HO-1 and SOCS-3 were induced in A549 cells and human pulmonary alveolar epithelial cells (HPAEpiCs) treated with (-)-epigallocatechin-3-gallate (EGCG). epigallocatechin gallate 134-164 heme oxygenase 1 Homo sapiens 21-25 22819551-3 2013 Here, we report that HO-1 and SOCS-3 were induced in A549 cells and human pulmonary alveolar epithelial cells (HPAEpiCs) treated with (-)-epigallocatechin-3-gallate (EGCG). epigallocatechin gallate 166-170 heme oxygenase 1 Homo sapiens 21-25 25506596-6 2013 Heme also induces a cytoprotective response that includes Nrf2 responsive genes such as heme oxygenase-1, ferritin, haptoglobin, hemopexin, and other antioxidant response genes. Heme 0-4 heme oxygenase 1 Homo sapiens 88-104 22945906-6 2013 Nicotine caused a dose-dependent decrease in cell proliferation, decreased heme oxygenase-1 (HO-1) expression (p < 0.05) and attenuated osteogenesis (p < 0.05) in hMSCs (45 % reduction at day 14). Nicotine 0-8 heme oxygenase 1 Homo sapiens 75-91 22945906-6 2013 Nicotine caused a dose-dependent decrease in cell proliferation, decreased heme oxygenase-1 (HO-1) expression (p < 0.05) and attenuated osteogenesis (p < 0.05) in hMSCs (45 % reduction at day 14). Nicotine 0-8 heme oxygenase 1 Homo sapiens 93-97 22945906-8 2013 Induction of HO-1 by peroxisome proliferator-activated receptor delta agonist (GW0742) prevented the effect of nicotine. GW0742 79-85 heme oxygenase 1 Homo sapiens 13-17 22945906-8 2013 Induction of HO-1 by peroxisome proliferator-activated receptor delta agonist (GW0742) prevented the effect of nicotine. Nicotine 111-119 heme oxygenase 1 Homo sapiens 13-17 22945906-10 2013 Therefore, induction of HO-1 prevents the deleterious effects of nicotine on osteogenesis in hMSC. Nicotine 65-73 heme oxygenase 1 Homo sapiens 24-28 22901690-6 2013 DHA activated Akt, p38 and ERK1/2 phosphorylation, and specific inhibitors of respective pathways attenuated DHA-induced Nrf2 nuclear translocation and HO-1 expression. Docosahexaenoic Acids 109-112 heme oxygenase 1 Homo sapiens 152-156 22901690-7 2013 Transfection with HO-1 siRNA knocked down HO-1 expression and partially reversed the DHA-mediated inhibition of TNF-alpha-induced p65 nuclear translocation and ICAM-1 expression. Docosahexaenoic Acids 85-88 heme oxygenase 1 Homo sapiens 18-22 22901690-8 2013 Importantly, we show for the first time that HO-1 plays a down-regulatory role in NF-kappaB nuclear translocation, and inhibition of Nrf2 ubiquitination and proteasome activity are involved in increased cellular Nrf2 level by DHA. Docosahexaenoic Acids 226-229 heme oxygenase 1 Homo sapiens 45-49 22901690-9 2013 In this study, we show that HO-1 plays a down-regulatory role in NF-kappaB nuclear translocation and that the protective effect of DHA against inflammation is partially via up-regulation of Nrf2-mediated HO-1 expression and inhibition of IKK/NF-kappaB signaling pathway. Docosahexaenoic Acids 131-134 heme oxygenase 1 Homo sapiens 204-208 22901690-4 2013 DHA triggered early-stage and transient reactive oxygen species (ROS) generation and significantly increased the protein expression of heme oxygenase 1 (HO-1), induced nuclear factor erythroid 2-related factor 2 (Nrf2) translocation to the nucleus and up-regulated antioxidant response element (ARE)-luciferase reporter activity. Docosahexaenoic Acids 0-3 heme oxygenase 1 Homo sapiens 135-151 22901690-4 2013 DHA triggered early-stage and transient reactive oxygen species (ROS) generation and significantly increased the protein expression of heme oxygenase 1 (HO-1), induced nuclear factor erythroid 2-related factor 2 (Nrf2) translocation to the nucleus and up-regulated antioxidant response element (ARE)-luciferase reporter activity. Docosahexaenoic Acids 0-3 heme oxygenase 1 Homo sapiens 153-157 22901690-6 2013 DHA activated Akt, p38 and ERK1/2 phosphorylation, and specific inhibitors of respective pathways attenuated DHA-induced Nrf2 nuclear translocation and HO-1 expression. Docosahexaenoic Acids 0-3 heme oxygenase 1 Homo sapiens 152-156 24101950-0 2013 Therapeutic roles of heme oxygenase-1 in metabolic diseases: curcumin and resveratrol analogues as possible inducers of heme oxygenase-1. Curcumin 61-69 heme oxygenase 1 Homo sapiens 120-136 23538677-3 2013 In this study, we investigated effects of fisetin, a natural bioactive flavonoid that has been reported to induce HO-1 expression, on the differentiation of macrophages into OCLs. fisetin 42-49 heme oxygenase 1 Homo sapiens 114-118 23864927-7 2013 SUL exerted antioxidant potential by upregulating expression of antioxidant enzymes including gamma-glutamylcysteine ligase, NAD(P)H:quinone oxidoreductase-1, and heme oxygenase-1 via activation of NF-E2-related factor 2(Nrf2). sulforaphane 0-3 heme oxygenase 1 Homo sapiens 163-179 24101950-0 2013 Therapeutic roles of heme oxygenase-1 in metabolic diseases: curcumin and resveratrol analogues as possible inducers of heme oxygenase-1. Resveratrol 74-85 heme oxygenase 1 Homo sapiens 120-136 24288584-10 2013 Thus, our results suggest that quercetin enhances mitochondrial biogenesis mainly via the HO-1/CO system in vitro and in vivo. Quercetin 31-40 heme oxygenase 1 Homo sapiens 90-97 23536765-2 2013 Since the heme catabolic pathway plays an important role in antioxidant protection, we attempted to assess the gene expression of key enzymes of heme catabolism, heme oxygenase 1 (HMOX1), heme oxygenase 2 (HMOX2), and biliverdin reductase A (BLVRA) in the liver and peripheral blood leukocytes (PBL) of patients chronically infected with HCV. Heme 10-14 heme oxygenase 1 Homo sapiens 162-178 23134284-10 2013 The expression of HO-1 and nuclear translocation of Nrf2 were increased by Abeta treatment. UNII-042A8N37WH 75-80 heme oxygenase 1 Homo sapiens 18-22 23536765-2 2013 Since the heme catabolic pathway plays an important role in antioxidant protection, we attempted to assess the gene expression of key enzymes of heme catabolism, heme oxygenase 1 (HMOX1), heme oxygenase 2 (HMOX2), and biliverdin reductase A (BLVRA) in the liver and peripheral blood leukocytes (PBL) of patients chronically infected with HCV. Heme 10-14 heme oxygenase 1 Homo sapiens 180-185 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. heme - carbon monoxide 105-127 heme oxygenase 1 Homo sapiens 11-27 23874015-4 2013 HO-1, a microsomal enzyme, catalyzes the breakdown of pro-oxidant heme, which is released from heme proteins to equimolar quantities of iron, carbon monoxide, and biliverdin. Heme 66-70 heme oxygenase 1 Homo sapiens 0-4 23874015-4 2013 HO-1, a microsomal enzyme, catalyzes the breakdown of pro-oxidant heme, which is released from heme proteins to equimolar quantities of iron, carbon monoxide, and biliverdin. Iron 136-140 heme oxygenase 1 Homo sapiens 0-4 23874015-4 2013 HO-1, a microsomal enzyme, catalyzes the breakdown of pro-oxidant heme, which is released from heme proteins to equimolar quantities of iron, carbon monoxide, and biliverdin. Carbon Monoxide 142-157 heme oxygenase 1 Homo sapiens 0-4 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. heme - carbon monoxide 105-127 heme oxygenase 1 Homo sapiens 29-33 23874015-4 2013 HO-1, a microsomal enzyme, catalyzes the breakdown of pro-oxidant heme, which is released from heme proteins to equimolar quantities of iron, carbon monoxide, and biliverdin. Biliverdine 163-173 heme oxygenase 1 Homo sapiens 0-4 23874015-6 2013 The beneficial effects of HO-1 expression are not merely due to heme degradation but are also attributed to the cytoprotective properties of the byproducts of the reaction. Heme 64-68 heme oxygenase 1 Homo sapiens 26-30 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Carbon Monoxide 129-131 heme oxygenase 1 Homo sapiens 11-27 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Carbon Monoxide 129-131 heme oxygenase 1 Homo sapiens 29-33 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Biliverdine 134-144 heme oxygenase 1 Homo sapiens 11-27 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Biliverdine 134-144 heme oxygenase 1 Homo sapiens 29-33 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Iron 150-154 heme oxygenase 1 Homo sapiens 11-27 23140858-2 2013 The enzyme heme oxygenase-1 (HO-1) generates three separate signaling molecules through the catalysis of heme - carbon monoxide (CO), biliverdin, and iron - each of which acts via distinct molecular targets to influence cell function, both proximally and distally. Iron 150-154 heme oxygenase 1 Homo sapiens 29-33 22444871-0 2012 Inhibition of NF-kappaB nuclear translocation via HO-1 activation underlies alpha-tocopheryl succinate toxicity. alpha-Tocopherol 76-102 heme oxygenase 1 Homo sapiens 50-54 22882835-0 2012 Evaluation of imidazole-based compounds as heme oxygenase-1 inhibitors. imidazole 14-23 heme oxygenase 1 Homo sapiens 43-59 22882835-4 2012 The binding of the most active compound 11 with heme or heme-conjugated human heme oxygenase-1 was also examined by spectral analyses. Heme 56-60 heme oxygenase 1 Homo sapiens 78-94 23217388-10 2012 The mRNA expression of HO-1 in the high glucose group was significantly increased in a time-dependent manner compared with that in the control group (P < 0.05). Glucose 40-47 heme oxygenase 1 Homo sapiens 23-27 23217388-11 2012 Adiponectin further increased the mRNA expression of HO-1 induced by high glucose in a time-dependent manner (P < 0.05).The expression of p66Shc was significantly increased in high glucose group compared with that in the control group (P < 0.05). Glucose 74-81 heme oxygenase 1 Homo sapiens 53-57 23217388-11 2012 Adiponectin further increased the mRNA expression of HO-1 induced by high glucose in a time-dependent manner (P < 0.05).The expression of p66Shc was significantly increased in high glucose group compared with that in the control group (P < 0.05). Glucose 184-191 heme oxygenase 1 Homo sapiens 53-57 23000247-6 2012 The CPN-9-mediated upregulation of HO-1, NQO1, and GCLM was abolished by Nrf2 knockdown. cpn-9 4-9 heme oxygenase 1 Homo sapiens 35-39 23100518-2 2012 This acquired neutrophil dysfunction is a consequence of induction of the cytoprotective, heme-degrading enzyme heme oxygenase-1 (HO-1) in neutrophil progenitors in bone marrow. Heme 90-94 heme oxygenase 1 Homo sapiens 112-128 23100518-2 2012 This acquired neutrophil dysfunction is a consequence of induction of the cytoprotective, heme-degrading enzyme heme oxygenase-1 (HO-1) in neutrophil progenitors in bone marrow. Heme 90-94 heme oxygenase 1 Homo sapiens 130-134 23144452-6 2012 Dietary CLA and nitrite supplementation in rodents elevates NO(2)-CLA levels in plasma, urine, and tissues, which in turn induces heme oxygenase-1 (HO-1) expression in the colonic epithelium. Linoleic Acids, Conjugated 8-11 heme oxygenase 1 Homo sapiens 130-146 23144452-6 2012 Dietary CLA and nitrite supplementation in rodents elevates NO(2)-CLA levels in plasma, urine, and tissues, which in turn induces heme oxygenase-1 (HO-1) expression in the colonic epithelium. Nitrites 16-23 heme oxygenase 1 Homo sapiens 130-146 23144452-6 2012 Dietary CLA and nitrite supplementation in rodents elevates NO(2)-CLA levels in plasma, urine, and tissues, which in turn induces heme oxygenase-1 (HO-1) expression in the colonic epithelium. Nitrogen Dioxide 60-65 heme oxygenase 1 Homo sapiens 130-146 22444871-6 2012 This effect is suppressed by the pharmacological inhibition of HO-1 and mimicked by the end-products of HO activity, i.e., bilirubin and carbon monoxide. Bilirubin 123-132 heme oxygenase 1 Homo sapiens 63-67 22956150-3 2012 Aim of investigation was to study the effects of cholinergic precursors (choline, CDP-choline, Acetylcholine and alpha-Glyceril-Phosphorylcholine) on HO-1 and p21 expression during astroglial cell proliferation and differentiation in primary cultures at 14 and 35 days in vitro (DIV) treated for 24 h with choline metabolites. Choline 49-56 heme oxygenase 1 Homo sapiens 150-154 22956150-3 2012 Aim of investigation was to study the effects of cholinergic precursors (choline, CDP-choline, Acetylcholine and alpha-Glyceril-Phosphorylcholine) on HO-1 and p21 expression during astroglial cell proliferation and differentiation in primary cultures at 14 and 35 days in vitro (DIV) treated for 24 h with choline metabolites. Acetylcholine 95-108 heme oxygenase 1 Homo sapiens 150-154 22956150-3 2012 Aim of investigation was to study the effects of cholinergic precursors (choline, CDP-choline, Acetylcholine and alpha-Glyceril-Phosphorylcholine) on HO-1 and p21 expression during astroglial cell proliferation and differentiation in primary cultures at 14 and 35 days in vitro (DIV) treated for 24 h with choline metabolites. alpha-glyceril-phosphorylcholine 113-145 heme oxygenase 1 Homo sapiens 150-154 22956150-3 2012 Aim of investigation was to study the effects of cholinergic precursors (choline, CDP-choline, Acetylcholine and alpha-Glyceril-Phosphorylcholine) on HO-1 and p21 expression during astroglial cell proliferation and differentiation in primary cultures at 14 and 35 days in vitro (DIV) treated for 24 h with choline metabolites. Choline 73-80 heme oxygenase 1 Homo sapiens 150-154 22956150-5 2012 On the contrary, ACh and choline induced a significant increase of HO-1 expression in 14 DIV astrocyte cultures. Acetylcholine 17-20 heme oxygenase 1 Homo sapiens 67-71 22956150-5 2012 On the contrary, ACh and choline induced a significant increase of HO-1 expression in 14 DIV astrocyte cultures. Choline 25-32 heme oxygenase 1 Homo sapiens 67-71 22956150-6 2012 Surprisingly, choline and ACh dramatically reduced HO-1 expression at 35 DIV. Choline 14-21 heme oxygenase 1 Homo sapiens 51-55 22956150-6 2012 Surprisingly, choline and ACh dramatically reduced HO-1 expression at 35 DIV. Acetylcholine 26-29 heme oxygenase 1 Homo sapiens 51-55 23171578-2 2012 CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Metals 29-34 heme oxygenase 1 Homo sapiens 97-113 23300448-10 2012 Moreover, HMOX-1 mRNA levels are enhanced in lovastatin-treated healthy and infected brains. Lovastatin 45-55 heme oxygenase 1 Homo sapiens 10-16 23022510-0 2012 L-ascorbate attenuates methamphetamine neurotoxicity through enhancing the induction of endogenous heme oxygenase-1. Ascorbic Acid 0-11 heme oxygenase 1 Homo sapiens 99-115 23022510-0 2012 L-ascorbate attenuates methamphetamine neurotoxicity through enhancing the induction of endogenous heme oxygenase-1. Methamphetamine 23-38 heme oxygenase 1 Homo sapiens 99-115 23171578-2 2012 CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Metals 29-34 heme oxygenase 1 Homo sapiens 115-120 23171578-2 2012 CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Heme 97-101 heme oxygenase 1 Homo sapiens 115-120 23171578-2 2012 CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Biliverdine 189-199 heme oxygenase 1 Homo sapiens 97-113 23171578-2 2012 CO-RMs containing transition metal carbonyls were initially implemented to mimic the function of heme oxygenase-1 (HMOX1), a stress inducible defensive protein that degrades heme to CO and biliverdin leading to anti-oxidant and anti-inflammatory actions. Biliverdine 189-199 heme oxygenase 1 Homo sapiens 115-120 22750196-4 2012 Induction of HO-1 during chronic hypoxia is necessary for the continued breakdown of heme for the enhanced production of hemoglobin and the increased respiratory and sympathetic responses. Heme 85-89 heme oxygenase 1 Homo sapiens 13-17 24009841-4 2012 Activation of Nrf2 by triterpenoids induces the expression of phase 2 detoxifying and antioxidant enzymes such as NAD(P)H quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1) - proteins which can protect cells or tissues against various toxic metabolites. triterpenoids 22-35 heme oxygenase 1 Homo sapiens 158-174 23026832-8 2012 However, LY294002 markedly quenched ROS and NO generation and diminished the protein expression of heme peroxidase enzyme (HO-1) and inducible nitric oxide synthase (iNOS). 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 9-17 heme oxygenase 1 Homo sapiens 123-127 22965812-2 2012 The expression of heme oxygenase-1 (HO-1), the rate-limiting enzyme involved in heme degradation, correlates well with the severity of psoriasis, and is a heritable trait. Heme 18-22 heme oxygenase 1 Homo sapiens 36-40 22965812-3 2012 This study aimed to assess the role of (GT)(n) dinucleotide repeat polymorphisms in the promoter region of the HO-1 gene in Chinese-Taiwanese patients with psoriasis. (n) dinucleotide 43-59 heme oxygenase 1 Homo sapiens 111-115 22966170-2 2012 Heme oxygenase-1 (gene HMOX1; protein HO-1) is the inducible, rate-limiting enzyme in the catabolism of heme and might attenuate the severity of outcomes from vaso-occlusive and hemolytic crises. Heme 104-108 heme oxygenase 1 Homo sapiens 0-16 22966170-2 2012 Heme oxygenase-1 (gene HMOX1; protein HO-1) is the inducible, rate-limiting enzyme in the catabolism of heme and might attenuate the severity of outcomes from vaso-occlusive and hemolytic crises. Heme 104-108 heme oxygenase 1 Homo sapiens 23-28 22966170-3 2012 A (GT)(n) dinucleotide repeat located in the promoter region of the HMOX1 gene is highly polymorphic, with long repeat lengths linked to decreased activity and inducibility. (n) dinucleotide 6-22 heme oxygenase 1 Homo sapiens 68-73 23187004-5 2012 Pretreatment with Ly294002, a PI3K inhibitor, augmented the decrease in HO-1 level by their combination, whereas no obvious changes were observed in Nrf2 levels. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 18-26 heme oxygenase 1 Homo sapiens 72-76 22982394-5 2012 In addition, isochlorogenic acid A declined markedly the content of hepatitis B virus covalently closed circular DNA (HBV cccDNA) and induced significantly the heme oxygenase-1 (HO-1) expression in HepG2.2.15 cells. Isochlorogenic acid A 13-34 heme oxygenase 1 Homo sapiens 178-182 22922731-0 2012 Curcumin inhibits HCV replication by induction of heme oxygenase-1 and suppression of AKT. Curcumin 0-8 heme oxygenase 1 Homo sapiens 50-66 22922731-5 2012 Under the same conditions, curcumin also dose-dependently induced heme oxygenase-1 with the highest induction at 24 h. Hemin, a heme oxygenase-1 inducer, also inhibited HCV protein expression in a dose-dependent manner. Curcumin 27-35 heme oxygenase 1 Homo sapiens 66-82 22922731-5 2012 Under the same conditions, curcumin also dose-dependently induced heme oxygenase-1 with the highest induction at 24 h. Hemin, a heme oxygenase-1 inducer, also inhibited HCV protein expression in a dose-dependent manner. Curcumin 27-35 heme oxygenase 1 Homo sapiens 128-144 22922731-6 2012 The knockdown of heme oxygenase-1 partially reversed the curcumin-inhibited HCV protein expression. Curcumin 57-65 heme oxygenase 1 Homo sapiens 17-33 22922731-7 2012 In addition to the heme oxygenase-1 induction, signaling molecule activities of AKT, extracellular signal-regulated kinases (ERK) and nuclear factor-kappaB (NF-kappaB) were inhibited by curcumin. Curcumin 186-194 heme oxygenase 1 Homo sapiens 19-35 22922731-10 2012 In summary, curcumin inhibited HCV replication by heme oxygenase-1 induction and AKT pathway inhibition. Curcumin 12-20 heme oxygenase 1 Homo sapiens 50-66 22940790-1 2012 In response to reactive oxygen species (ROS) or electrophiles, the transcription factor nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) rapidly translocates into the nucleus and induces the expression of various antioxidant genes, such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 15-38 heme oxygenase 1 Homo sapiens 250-266 22940790-1 2012 In response to reactive oxygen species (ROS) or electrophiles, the transcription factor nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) rapidly translocates into the nucleus and induces the expression of various antioxidant genes, such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 15-38 heme oxygenase 1 Homo sapiens 268-272 22940790-1 2012 In response to reactive oxygen species (ROS) or electrophiles, the transcription factor nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) rapidly translocates into the nucleus and induces the expression of various antioxidant genes, such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 40-43 heme oxygenase 1 Homo sapiens 250-266 22940790-1 2012 In response to reactive oxygen species (ROS) or electrophiles, the transcription factor nuclear factor-erythroid 2 (NF-E2)-related factor 2 (Nrf2) rapidly translocates into the nucleus and induces the expression of various antioxidant genes, such as heme oxygenase-1 (HO-1). Reactive Oxygen Species 40-43 heme oxygenase 1 Homo sapiens 268-272 22982394-8 2012 Overexpression of HO-1 may contribute to the anti-HBV activity of isochlorogenic acid A by reducing the stability of the HBV core protein and thus blocking the refill of nuclear HBV cccDNA. Isochlorogenic acid A 66-87 heme oxygenase 1 Homo sapiens 18-22 22982394-9 2012 Additionally, the hepatoprotective effect of isochlorogenic acid A could be achieved by its antioxidative property and induction of HO-1. Isochlorogenic acid A 45-66 heme oxygenase 1 Homo sapiens 132-136 22864061-2 2012 In this study, we investigated whether sildenafil promotes the production of the sGC-stimulatory gases, carbon monoxide and nitric oxide, by stimulating the expression of the inducible isoforms of heme oxygenase (HO-1) and nitric oxide synthase (iNOS) in vascular smooth muscle cells (SMCs). Sildenafil Citrate 39-49 heme oxygenase 1 Homo sapiens 213-217 22659318-0 2012 Cadmium-induced apoptosis in the BJAB human B cell line: involvement of PKC/ERK1/2/JNK signaling pathways in HO-1 expression. Cadmium 0-7 heme oxygenase 1 Homo sapiens 109-113 22659318-5 2012 The objective of the present study was to investigate the mechanism of HO-1 induction during cadmium (Cd)-induced oxidative stress and apoptosis in the lymphocyte B cell line BJAB. Cadmium 93-100 heme oxygenase 1 Homo sapiens 71-75 22659318-5 2012 The objective of the present study was to investigate the mechanism of HO-1 induction during cadmium (Cd)-induced oxidative stress and apoptosis in the lymphocyte B cell line BJAB. Cadmium 102-104 heme oxygenase 1 Homo sapiens 71-75 22659318-8 2012 Our results indicate that Cd also induces HO-1 expression which is modulated by the thiol redox status, tyrosine kinase and PI3-kinase. Cadmium 26-28 heme oxygenase 1 Homo sapiens 42-46 22659318-8 2012 Our results indicate that Cd also induces HO-1 expression which is modulated by the thiol redox status, tyrosine kinase and PI3-kinase. Sulfhydryl Compounds 84-89 heme oxygenase 1 Homo sapiens 42-46 22659318-9 2012 The inhibitory effect of calphostin C suggests that Cd induction of HO-1 expression could be mediated by the PKC pathway in the BJAB cells together with the involvement of ERK 1/2 and JNK in a dose-dependent manner. Cadmium 52-54 heme oxygenase 1 Homo sapiens 68-72 22864061-3 2012 Sildenafil increased HO-1 expression and potentiated cytokine-mediated expression of iNOS and NO synthesis by SMCs. Sildenafil Citrate 0-10 heme oxygenase 1 Homo sapiens 21-25 22864061-5 2012 However, the sildenafil-mediated increase in HO-1 promoter activity was abolished by mutating the antioxidant responsive elements in the promoter or by overexpressing a dominant-negative mutant of NF-E2-related factor-2 (Nrf2). Sildenafil Citrate 13-23 heme oxygenase 1 Homo sapiens 45-49 22842631-0 2012 Globular adiponectin inhibits ethanol-induced apoptosis in HepG2 cells through heme oxygenase-1 induction. Ethanol 30-37 heme oxygenase 1 Homo sapiens 79-95 22864061-6 2012 Furthermore, the induction of HO-1 by sildenafil was accompanied by an increase in reactive oxygen species (ROS) and blocked by N-acetyl-L-cysteine and rotenone. Sildenafil Citrate 38-48 heme oxygenase 1 Homo sapiens 30-34 22864061-6 2012 Furthermore, the induction of HO-1 by sildenafil was accompanied by an increase in reactive oxygen species (ROS) and blocked by N-acetyl-L-cysteine and rotenone. Reactive Oxygen Species 83-106 heme oxygenase 1 Homo sapiens 30-34 22864061-6 2012 Furthermore, the induction of HO-1 by sildenafil was accompanied by an increase in reactive oxygen species (ROS) and blocked by N-acetyl-L-cysteine and rotenone. Reactive Oxygen Species 108-111 heme oxygenase 1 Homo sapiens 30-34 22864061-6 2012 Furthermore, the induction of HO-1 by sildenafil was accompanied by an increase in reactive oxygen species (ROS) and blocked by N-acetyl-L-cysteine and rotenone. Acetylcysteine 128-147 heme oxygenase 1 Homo sapiens 30-34 22864061-6 2012 Furthermore, the induction of HO-1 by sildenafil was accompanied by an increase in reactive oxygen species (ROS) and blocked by N-acetyl-L-cysteine and rotenone. Rotenone 152-160 heme oxygenase 1 Homo sapiens 30-34 22864061-8 2012 In conclusion, these studies demonstrate that sildenafil stimulates the expression of HO-1 and iNOS via the ROS-Nrf2 and sGC-cGMP pathway, respectively. Sildenafil Citrate 46-56 heme oxygenase 1 Homo sapiens 86-90 22864061-8 2012 In conclusion, these studies demonstrate that sildenafil stimulates the expression of HO-1 and iNOS via the ROS-Nrf2 and sGC-cGMP pathway, respectively. Reactive Oxygen Species 108-111 heme oxygenase 1 Homo sapiens 86-90 22864061-8 2012 In conclusion, these studies demonstrate that sildenafil stimulates the expression of HO-1 and iNOS via the ROS-Nrf2 and sGC-cGMP pathway, respectively. Cyclic GMP 125-129 heme oxygenase 1 Homo sapiens 86-90 22864061-9 2012 The ability of sildenafil to block the catabolism of cGMP while stimulating the synthesis of sGC-stimulatory gaseous monoxides through the induction of HO-1 and iNOS provides a potent mechanism by which cGMP-dependent vascular actions of this drug are amplified. Sildenafil Citrate 15-25 heme oxygenase 1 Homo sapiens 152-156 22864061-9 2012 The ability of sildenafil to block the catabolism of cGMP while stimulating the synthesis of sGC-stimulatory gaseous monoxides through the induction of HO-1 and iNOS provides a potent mechanism by which cGMP-dependent vascular actions of this drug are amplified. monoxides 117-126 heme oxygenase 1 Homo sapiens 152-156 22864061-9 2012 The ability of sildenafil to block the catabolism of cGMP while stimulating the synthesis of sGC-stimulatory gaseous monoxides through the induction of HO-1 and iNOS provides a potent mechanism by which cGMP-dependent vascular actions of this drug are amplified. Cyclic GMP 203-207 heme oxygenase 1 Homo sapiens 152-156 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Heme 6-10 heme oxygenase 1 Homo sapiens 31-44 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Oxygen 11-17 heme oxygenase 1 Homo sapiens 31-44 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Iron 140-144 heme oxygenase 1 Homo sapiens 6-29 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Iron 140-144 heme oxygenase 1 Homo sapiens 31-44 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Carbon Monoxide 146-148 heme oxygenase 1 Homo sapiens 6-29 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Carbon Monoxide 146-148 heme oxygenase 1 Homo sapiens 31-44 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Biliverdine 154-164 heme oxygenase 1 Homo sapiens 6-29 22923613-1 2012 Human heme oxygenases 1 and 2 (HO-1 and HO-2) degrade heme in the presence of oxygen and NADPH-cytochrome P450 reductase, producing ferrous iron, CO, and biliverdin. Biliverdine 154-164 heme oxygenase 1 Homo sapiens 31-44 22875631-0 2012 The heme oxygenase-1 inhibitor ZnPPIX induces non-canonical, Beclin 1-independent, autophagy through p38 MAPK pathway. zinc protoporphyrin 31-37 heme oxygenase 1 Homo sapiens 4-20 22875631-1 2012 Zinc protoporphyrin IX (ZnPPIX), a heme oxygenase-1 enzyme inhibitor, has been reported to induce apoptosis and to have antitumor properties. zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 35-51 22875631-1 2012 Zinc protoporphyrin IX (ZnPPIX), a heme oxygenase-1 enzyme inhibitor, has been reported to induce apoptosis and to have antitumor properties. zinc protoporphyrin 24-30 heme oxygenase 1 Homo sapiens 35-51 22954673-4 2012 Additionally, heme activates redox-sensitive proliferation-related signaling routes, such as mitogen activated protein kinase (MAPK) and NF-kappaB, and induces heme oxygenase-1 (HO-1) expression. Heme 14-18 heme oxygenase 1 Homo sapiens 160-176 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. gacrp 157-162 heme oxygenase 1 Homo sapiens 75-79 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. gacrp 157-162 heme oxygenase 1 Homo sapiens 75-79 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. gacrp 235-240 heme oxygenase 1 Homo sapiens 75-79 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. gacrp 235-240 heme oxygenase 1 Homo sapiens 75-79 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. Ethanol 244-251 heme oxygenase 1 Homo sapiens 75-79 22842631-8 2012 Treatment with SnPP, a pharmacological inhibitor of HO-1, and knockdown of HO-1 with small interfering RNA (siRNA) restored caspase-3 activity suppressed by gAcrp, indicating a critical role of HO-1 in mediating the protective role of gAcrp in ethanol-induced apoptosis in liver cells. Ethanol 244-251 heme oxygenase 1 Homo sapiens 75-79 22842631-10 2012 In conclusion, these data demonstrated that globular adiponectin prevents ethanol-induced apoptosis in HepG2 cells via HO-1 induction and revealed a novel biological response of globular adiponectin in the protection of liver injury from alcohol consumption. Ethanol 74-81 heme oxygenase 1 Homo sapiens 119-123 22828666-8 2012 At a higher dose, 20 mug/ml, carnosic acid activated the expression of antioxidant (AKR1C2, TNXRD1, HMOX1) and apoptosis (GDF15, PHLDA1, DDIT3) genes and suppressed the expression of inhibitor of transcription (ID3) and cell cycle (CDKN2C) genes. salvin 29-42 heme oxygenase 1 Homo sapiens 100-105 22801788-3 2012 Vitamin D also exerts protective effects against SLE through non-genomic factors, such as ultraviolet radiation (UV) exposure, matrix metalloproteinase (MMPs), heme oxygenase-1 (HO-1), the prostaglandins (PGs), cyclooxygenase-2 (COX-2), and oxidative stress. Vitamin D 0-9 heme oxygenase 1 Homo sapiens 160-176 22777293-4 2012 SUMMARY: Treatments based on direct heme-mimetics or other agonists of this pathway have enormous potential for linked antioxidant protection via heme oxygenase 1 and reduced foam cell formation via liver X receptor, a potent combination for treating atherosclerosis. Heme 36-40 heme oxygenase 1 Homo sapiens 146-162 22212955-4 2012 The (GT)(n) and (TA)(n) dinucleotide variations in HMOX1 and UGT1A1 gene promoters, respectively, were determined by fragment analysis. (n) dinucleotide 21-38 heme oxygenase 1 Homo sapiens 53-58 22212955-0 2012 Association of serum bilirubin and promoter variations in HMOX1 and UGT1A1 genes with sporadic colorectal cancer. Bilirubin 21-30 heme oxygenase 1 Homo sapiens 58-63 22419571-1 2012 Heme-oxygenase 1 is an endoplasmic reticulum-anchored enzyme that breaks down heme into iron, carbon monoxide and biliverdin. Heme 78-82 heme oxygenase 1 Homo sapiens 0-16 22419571-1 2012 Heme-oxygenase 1 is an endoplasmic reticulum-anchored enzyme that breaks down heme into iron, carbon monoxide and biliverdin. Iron 88-92 heme oxygenase 1 Homo sapiens 0-16 22419571-1 2012 Heme-oxygenase 1 is an endoplasmic reticulum-anchored enzyme that breaks down heme into iron, carbon monoxide and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 0-16 22419571-1 2012 Heme-oxygenase 1 is an endoplasmic reticulum-anchored enzyme that breaks down heme into iron, carbon monoxide and biliverdin. Biliverdine 114-124 heme oxygenase 1 Homo sapiens 0-16 22881289-2 2012 The stress protein, HO-1 mediates the degradation of cellular heme to biliverdin/bilirubin, free iron, and CO and is up-regulated in the brains of persons with Alzheimer"s disease and Parkinson"s disease. Heme 62-66 heme oxygenase 1 Homo sapiens 20-24 22227268-5 2012 Lico-E activates Nrf2-antioxidant response element (ARE) system and up-regulates downstream NAD(P)H:quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1). licochalcone E 0-6 heme oxygenase 1 Homo sapiens 154-158 22227268-6 2012 Anti-inflammatory and cytoprotective effects of Lico-E are attenuated in siRNA-mediated Nrf2-silencing cells as well as in the presence with specific inhibitor of HO-1 or NQO1, respectively. licochalcone E 48-54 heme oxygenase 1 Homo sapiens 163-167 23248504-1 2012 BACKGROUND: At low concentrations, carbon monoxide (CO) can confer cyto and tissue-protective effects, such as endogenous Heme oxygenase 1 expression, which has antioxidative, anti-inflammatory, antiproliferative, and antiapoptotic effects. Carbon Monoxide 35-50 heme oxygenase 1 Homo sapiens 122-138 23248504-1 2012 BACKGROUND: At low concentrations, carbon monoxide (CO) can confer cyto and tissue-protective effects, such as endogenous Heme oxygenase 1 expression, which has antioxidative, anti-inflammatory, antiproliferative, and antiapoptotic effects. Carbon Monoxide 52-54 heme oxygenase 1 Homo sapiens 122-138 22881289-2 2012 The stress protein, HO-1 mediates the degradation of cellular heme to biliverdin/bilirubin, free iron, and CO and is up-regulated in the brains of persons with Alzheimer"s disease and Parkinson"s disease. Biliverdine 70-80 heme oxygenase 1 Homo sapiens 20-24 22881289-2 2012 The stress protein, HO-1 mediates the degradation of cellular heme to biliverdin/bilirubin, free iron, and CO and is up-regulated in the brains of persons with Alzheimer"s disease and Parkinson"s disease. Bilirubin 81-90 heme oxygenase 1 Homo sapiens 20-24 22881289-2 2012 The stress protein, HO-1 mediates the degradation of cellular heme to biliverdin/bilirubin, free iron, and CO and is up-regulated in the brains of persons with Alzheimer"s disease and Parkinson"s disease. Iron 97-101 heme oxygenase 1 Homo sapiens 20-24 23302671-0 2012 [Effects of policosanol on serum lipids and heme oxygenase-1 in patients with hyperlipidemia]. policosanol 12-23 heme oxygenase 1 Homo sapiens 44-60 21756955-8 2012 Pretreatment of the astrocytes with antioxidants or a specific calcium chelator BAPTA-AM, significantly blocked the upregulation of Nrf2, HO-1 and Nqo1. Calcium 63-70 heme oxygenase 1 Homo sapiens 138-142 21756955-8 2012 Pretreatment of the astrocytes with antioxidants or a specific calcium chelator BAPTA-AM, significantly blocked the upregulation of Nrf2, HO-1 and Nqo1. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 80-88 heme oxygenase 1 Homo sapiens 138-142 23248671-8 2012 Pretreatment with L-serine (0.1-3.2 mM) protected endothelial cells from hydrogen peroxide-mediated cell cytotoxicity (H(2)O(2), 0.5 mM) and lead to significant induction of Nrf2 activity, HO-1 expresssion and NOx production. Serine 18-26 heme oxygenase 1 Homo sapiens 189-193 23248671-9 2012 These findings demonstrated that L-serine has antioxidant and cytoprotective effects through the elevation of some crucial antioxidant factors such as Nrf2, HO-1 and NO. Serine 33-41 heme oxygenase 1 Homo sapiens 157-168 22879597-2 2012 Cobalt protoporphyrin (CoPP), a well known heme oxygenase 1 inducer, has been used to promote endogenous CO generation and protect against ischemia/reperfusion injury. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 43-59 22879597-2 2012 Cobalt protoporphyrin (CoPP), a well known heme oxygenase 1 inducer, has been used to promote endogenous CO generation and protect against ischemia/reperfusion injury. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 43-59 23302671-1 2012 OBJECTIVE: To evaluate the effects of policosanol on serum lipid and heme oxygenase-1 (HO-1) in patients with hyperlipidemia. policosanol 38-49 heme oxygenase 1 Homo sapiens 69-85 23302671-1 2012 OBJECTIVE: To evaluate the effects of policosanol on serum lipid and heme oxygenase-1 (HO-1) in patients with hyperlipidemia. policosanol 38-49 heme oxygenase 1 Homo sapiens 87-91 22918643-10 2012 Simvastatin-dependent HO-1 protein induction was reduced significantly by pharmacological inhibition of the phosphotidylinositol-3-kinase (PI3K)/Akt pathways. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 22-26 22918643-12 2012 CONCLUSIONS: These results demonstrate that HO-1 is a target site and an antioxidant mediator of simvastatin in human RPE cells. Simvastatin 97-108 heme oxygenase 1 Homo sapiens 44-48 22918643-0 2012 Effects of simvastatin on the expression of heme oxygenase-1 in human RPE cells. Simvastatin 11-22 heme oxygenase 1 Homo sapiens 44-60 22918643-8 2012 Simvastatin increased the HO-1 mRNA and protein levels in a concentration-dependent manner up to 10 muM. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 26-30 22918643-13 2012 Simvastatin-dependent upregulation of HO-1 is mainly via PI3K/Akt-dependent signaling pathways. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 38-42 22918643-9 2012 HO-1 protein induction by simvastatin was unaffected by mevalonate or N-nitro-L-arginine methyl ester, showing that the isoprenoid- and NO-dependent pathways are not involved. Simvastatin 26-37 heme oxygenase 1 Homo sapiens 0-4 22917016-5 2012 Therefore, the molecular signaling events affecting resveratrol-mediated heme oxygenase-1 (HO-1) and glyoxalase expression levels were further investigated in this study. Resveratrol 52-63 heme oxygenase 1 Homo sapiens 73-89 22917016-5 2012 Therefore, the molecular signaling events affecting resveratrol-mediated heme oxygenase-1 (HO-1) and glyoxalase expression levels were further investigated in this study. Resveratrol 52-63 heme oxygenase 1 Homo sapiens 91-95 22917016-6 2012 Our findings indicated that resveratrol activated the extracellular signal-regulated kinase (ERK) pathway but not the p38 or c-Jun N-terminal kinase (JNK) pathways, subsequently leading to Nrf2 nuclear translocation and elevation of HO-1 and glyoxalase expression levels. Resveratrol 28-39 heme oxygenase 1 Homo sapiens 233-237 22917016-8 2012 In contrast, depletion of Nrf2 by small interfering RNA (si-RNA) resulted in the abrogation of HO-1 and glyoxalase expression in the MG-treated resveratrol group in Hep G2 cells. Resveratrol 144-155 heme oxygenase 1 Homo sapiens 95-99 22821808-0 2012 Tunicamycin inhibits PDGF-BB-induced proliferation and migration of vascular smooth muscle cells through induction of HO-1. Tunicamycin 0-11 heme oxygenase 1 Homo sapiens 118-122 22564156-3 2012 Hence, the ability of exogenously-added copper, iron and zinc to influence HO-1 expression in HCT-116 cells was evaluated. Copper 40-46 heme oxygenase 1 Homo sapiens 75-79 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sirolimus 0-9 heme oxygenase 1 Homo sapiens 105-109 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sirolimus 0-9 heme oxygenase 1 Homo sapiens 228-232 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sirolimus 11-15 heme oxygenase 1 Homo sapiens 105-109 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sirolimus 11-15 heme oxygenase 1 Homo sapiens 228-232 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sorafenib 21-30 heme oxygenase 1 Homo sapiens 105-109 22843690-5 2012 Rapamycin (RAPA) and sorafenib, two commonly used drugs for renal cancer treatment, were found to induce HO-1 expression in renal cancer cells Caki-1 and 786-O; and the apoptotic effect of these drugs was markedly enhanced upon HO-1 knockdown. Sorafenib 21-30 heme oxygenase 1 Homo sapiens 228-232 22843690-6 2012 Overexpression of HO-1 protected the cells from RAPA- and sorafenib-induced apoptosis and also averted drug-mediated inhibition of cell proliferation. Sirolimus 48-52 heme oxygenase 1 Homo sapiens 18-22 22843690-6 2012 Overexpression of HO-1 protected the cells from RAPA- and sorafenib-induced apoptosis and also averted drug-mediated inhibition of cell proliferation. Sorafenib 58-67 heme oxygenase 1 Homo sapiens 18-22 22709784-7 2012 Silymarin acted as a potent cytoprotective effector through the effective induction of anti-oxidant related genes, including Nrf2, NQO1 and HO-1, in the presence of PQ. Silymarin 0-9 heme oxygenase 1 Homo sapiens 140-144 22564156-2 2012 Toxic heavy metals induce HO-1, but it is unclear whether particular metal micronutrients also induce HO-1. Metals 12-17 heme oxygenase 1 Homo sapiens 26-30 22564156-3 2012 Hence, the ability of exogenously-added copper, iron and zinc to influence HO-1 expression in HCT-116 cells was evaluated. Iron 48-52 heme oxygenase 1 Homo sapiens 75-79 22564156-7 2012 In contrast, deferoxamine blunted the induction of HO-1 mRNA, protein, and enzymatic activity caused by zinc. Deferoxamine 13-25 heme oxygenase 1 Homo sapiens 51-55 22564156-8 2012 Additionally, N-acetylcysteine and Tiron inhibited zinc-induced HO-1 upregulation and also nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). Acetylcysteine 14-30 heme oxygenase 1 Homo sapiens 64-68 22564156-8 2012 Additionally, N-acetylcysteine and Tiron inhibited zinc-induced HO-1 upregulation and also nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt 35-40 heme oxygenase 1 Homo sapiens 64-68 22564156-9 2012 Collectively, these findings suggest that zinc at above normal levels upregulates HO-1 expression in HCT-116 cells in a ROS-independent manner. Reactive Oxygen Species 120-123 heme oxygenase 1 Homo sapiens 82-86 22583557-4 2012 After undergoing a partial catabolism, alkenals enter into a great number of body"s cells, where they react with Nrf2-Keap1 protein: the transfer of activated Nrf2 into the nucleus and its binding to antioxidant response element (ARE) is the crucial event able to upregulate the synthesis of antioxidant proteins, phase II enzymes and HO-1. alkenals 39-47 heme oxygenase 1 Homo sapiens 335-339 22137262-0 2012 Epigallocatechin gallate induces expression of heme oxygenase-1 in endothelial cells via p38 MAPK and Nrf-2 that suppresses proinflammatory actions of TNF-alpha. epigallocatechin gallate 0-24 heme oxygenase 1 Homo sapiens 47-63 22137262-3 2012 When compared with vehicle-treated controls, EGCG treatment (2.5 muM, 8 h) of human aortic endothelial cells (HAEC) caused a ~three-fold increase in heme oxygenase-1 (HO-1) mRNA and protein with comparable increases in HO-1 activity. epigallocatechin gallate 45-49 heme oxygenase 1 Homo sapiens 149-165 22487967-0 2012 15-deoxy-Delta12,14 -prostaglandin J2 inhibits human immunodeficiency virus-1 tat-induced monocyte chemoattractant protein-1/CCL2 production by blocking the extracellular signal-regulated kinase-1/2 signaling pathway independently of peroxisome proliferator-activated receptor-gamma and heme oxygenase-1 in rat hippocampal slices. 14 -prostaglandin j2 17-37 heme oxygenase 1 Homo sapiens 287-303 22137262-3 2012 When compared with vehicle-treated controls, EGCG treatment (2.5 muM, 8 h) of human aortic endothelial cells (HAEC) caused a ~three-fold increase in heme oxygenase-1 (HO-1) mRNA and protein with comparable increases in HO-1 activity. epigallocatechin gallate 45-49 heme oxygenase 1 Homo sapiens 167-171 22487967-7 2012 Despite 15d-PGJ2"s increasing the expression of heme oxygenase-1 (HO-1), its action was not abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX), nor was it recapitulated by HO-1 inducers such as cobalt protoporphyrin (CoPP). 15-deoxy-delta(12,14)-prostaglandin J2 8-16 heme oxygenase 1 Homo sapiens 48-64 22137262-3 2012 When compared with vehicle-treated controls, EGCG treatment (2.5 muM, 8 h) of human aortic endothelial cells (HAEC) caused a ~three-fold increase in heme oxygenase-1 (HO-1) mRNA and protein with comparable increases in HO-1 activity. epigallocatechin gallate 45-49 heme oxygenase 1 Homo sapiens 219-223 22487967-7 2012 Despite 15d-PGJ2"s increasing the expression of heme oxygenase-1 (HO-1), its action was not abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPPIX), nor was it recapitulated by HO-1 inducers such as cobalt protoporphyrin (CoPP). 15-deoxy-delta(12,14)-prostaglandin J2 8-16 heme oxygenase 1 Homo sapiens 66-70 22137262-5 2012 Pretreatment of HAEC with SB203580 (p38 MAPK inhibitor) or siRNA knockdown of p38 MAPK completely blocked EGCG-stimulated induction of HO-1. SB 203580 26-34 heme oxygenase 1 Homo sapiens 135-139 22487967-11 2012 Collectively, these data demonstrate that the antiinflammatory effects of 15d-PGJ2 on the hippocampus are exerted through inhibition of Tat-mediated ERK1/2 activation, coupled with that of a redox-sensitive pathway, independent of PPARgamma and HO-1. 15-deoxy-delta(12,14)-prostaglandin J2 74-82 heme oxygenase 1 Homo sapiens 245-249 22137262-5 2012 Pretreatment of HAEC with SB203580 (p38 MAPK inhibitor) or siRNA knockdown of p38 MAPK completely blocked EGCG-stimulated induction of HO-1. epigallocatechin gallate 106-110 heme oxygenase 1 Homo sapiens 135-139 22137262-7 2012 siRNA knockdown of HO-1, p38 MAPK or Nrf-2 blocked these inhibitory actions of EGCG. epigallocatechin gallate 79-83 heme oxygenase 1 Homo sapiens 19-23 22137262-8 2012 In HAEC transiently transfected with a human HO-1 promoter luciferase reporter (or an isolated Nrf-2 responsive region), luciferase activity increased in response to EGCG. epigallocatechin gallate 166-170 heme oxygenase 1 Homo sapiens 45-49 22137262-10 2012 EGCG-stimulated expression of HO-1 and Nrf-2 was blocked by siRNA knockdown of Nrf-2 or p38 MAPK. epigallocatechin gallate 0-4 heme oxygenase 1 Homo sapiens 30-34 22137262-11 2012 Finally, liver from mice chronically treated with EGCG had increased HO-1 and decreased VCAM-1 expression. epigallocatechin gallate 50-54 heme oxygenase 1 Homo sapiens 69-73 22137262-12 2012 Thus, in vascular endothelium, EGCG requires p38 MAPK to increase expression of Nrf-2 that drives expression of HO-1, resulting in increased HO-1 activity. epigallocatechin gallate 31-35 heme oxygenase 1 Homo sapiens 112-116 22137262-12 2012 Thus, in vascular endothelium, EGCG requires p38 MAPK to increase expression of Nrf-2 that drives expression of HO-1, resulting in increased HO-1 activity. epigallocatechin gallate 31-35 heme oxygenase 1 Homo sapiens 141-145 22137262-13 2012 Increased HO-1 expression may underlie anti-inflammatory actions of EGCG in vascular endothelium that may help mediate beneficial cardiovascular actions of green tea. epigallocatechin gallate 68-72 heme oxygenase 1 Homo sapiens 10-14 22819264-1 2012 INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). Heme 99-103 heme oxygenase 1 Homo sapiens 14-30 22819264-1 2012 INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). Biliverdine 109-119 heme oxygenase 1 Homo sapiens 14-30 22819264-1 2012 INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). Iron 126-130 heme oxygenase 1 Homo sapiens 14-30 22836558-7 2012 Free Hb binds to haptoglobin (Hp) and once Hp-Hb complex is endocytosed by CD163, liberated heme is converted into less toxic compounds by heme oxygenase-1. Heme 92-96 heme oxygenase 1 Homo sapiens 139-155 22819264-1 2012 INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). Carbon Monoxide 136-151 heme oxygenase 1 Homo sapiens 14-30 22819264-1 2012 INTRODUCTION: Heme oxygenase-1 (HO-1) is the rate limiting enzyme that catalyzes the conversion of heme into biliverdin, free iron, and carbon monoxide (CO). Carbon Monoxide 153-155 heme oxygenase 1 Homo sapiens 14-30 22843070-4 2012 Immunostaining and quantitative RT-PCR revealed that PQQ treatment promotes nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2), and up-regulates mRNA expression of Nrf2 and the antioxidant enzyme genes, heme oxygenase-1 and glutamate cysteine ligase catalytic in glutamate-injured hippocampal neurons; this is a process dependent on the PI3K/Akt pathway, as evidenced by blocking experiments with PI3K inhibitors. PQQ Cofactor 54-57 heme oxygenase 1 Homo sapiens 231-247 22735309-0 2012 Phenylpropanoid glycosides from plant cell cultures induce heme oxygenase 1 gene expression in a human keratinocyte cell line by affecting the balance of NRF2 and BACH1 transcription factors. phenylpropanoid glycosides 0-26 heme oxygenase 1 Homo sapiens 59-75 23158494-3 2012 Morphologic observations, flow cytometry Annexin-V and DNA analysis were used to detect apoptosis and determine the content of DNA of MSCs after a 12-hour treatment of Znpp-IX, a Ho-1 special inhibitor. zinc protoporphyrin 168-175 heme oxygenase 1 Homo sapiens 179-183 22078849-4 2012 However, no study that observed HO-1 in both epithelial and subepithelial tissues of CRS has been reported. 3-cresol 85-88 heme oxygenase 1 Homo sapiens 32-36 22901679-9 2012 Other parameters such as LDH and HO-1 were not influenced by gaseous compounds: Following aerosol exposure, LDH levels appeared elevated at both timepoints and the HO-1 transcript correlated positively with deposited ZnO-dose. Zinc Oxide 217-220 heme oxygenase 1 Homo sapiens 164-168 22891172-0 2012 Letter by Vitek et al regarding article, "niacin inhibits vascular inflammation via the induction of heme oxygenase-1". Niacin 42-48 heme oxygenase 1 Homo sapiens 101-118 21695420-6 2012 WI-38 fibroblasts exposed to a subcytotoxic concentration of copper sulfate presented inhibition of cell proliferation, cell enlargement, increased SA beta-gal activity, and mRNA overexpression of several senescence-associated genes such as p21, apolipoprotein J (ApoJ), fibronectin, transforming growth factor beta-1 (TGF beta1), insulin growth factor binding protein 3, and heme oxygenase 1. Copper Sulfate 61-75 heme oxygenase 1 Homo sapiens 376-392 22078849-6 2012 RESULTS: We found that the expression of HO-1 in the epithelial layers of CRS without eosinophilic infiltration was significantly enhanced as compared with that of CRS with eosinophilic infiltration. 3-cresol 74-77 heme oxygenase 1 Homo sapiens 41-45 22078849-6 2012 RESULTS: We found that the expression of HO-1 in the epithelial layers of CRS without eosinophilic infiltration was significantly enhanced as compared with that of CRS with eosinophilic infiltration. 3-cresol 164-167 heme oxygenase 1 Homo sapiens 41-45 22078849-7 2012 On the other hand, the number of macrophages with HO-1 positive reactions was significantly greater in CRS with eosinophilic infiltration compared with CRS without eosinophilic infiltration. 3-cresol 103-106 heme oxygenase 1 Homo sapiens 50-54 22078849-7 2012 On the other hand, the number of macrophages with HO-1 positive reactions was significantly greater in CRS with eosinophilic infiltration compared with CRS without eosinophilic infiltration. 3-cresol 152-155 heme oxygenase 1 Homo sapiens 50-54 22579918-0 2012 Sustained expression of heme oxygenase-1 alters iron homeostasis in nonerythroid cells. Iron 48-52 heme oxygenase 1 Homo sapiens 24-40 21547369-5 2012 Expression array analysis revealed DHA-induced upregulation of oxidative and genotoxic stress response genes (GADD45A, GADD153, CDKN1A, PMAIP1, HMOX1, EGR1) in A375 cells. artenimol 35-38 heme oxygenase 1 Homo sapiens 144-149 22683349-9 2012 Guanosine induced the antioxidant enzyme heme oxygenase-1 (HO-1) expression. Guanosine 0-9 heme oxygenase 1 Homo sapiens 41-57 22683349-9 2012 Guanosine induced the antioxidant enzyme heme oxygenase-1 (HO-1) expression. Guanosine 0-9 heme oxygenase 1 Homo sapiens 59-63 22683349-10 2012 The protective effects of guanosine were prevented by heme oxygenase-1 inhibitor, SnPP. Guanosine 26-35 heme oxygenase 1 Homo sapiens 54-70 22683349-11 2012 Moreover, bilirubin, an antioxidant and physiologic product of HO-1, is protective against mitochondrial oxidative stress. Bilirubin 10-19 heme oxygenase 1 Homo sapiens 63-67 22683349-12 2012 In conclusion, our results show that guanosine can afford protection against mitochondrial oxidative stress by a signaling pathway that implicates PI3K/Akt/GSK-3beta proteins and induction of the antioxidant enzyme HO-1. Guanosine 37-46 heme oxygenase 1 Homo sapiens 215-219 22484158-7 2012 Interestingly, the profound up-regulation of HO-1 and Nrf-2 were observed in only ethanol-challenged cells, which evidenced that lucidone-induced induction of HO-/Nrf-2 were specific with oxidative stress. Ethanol 82-89 heme oxygenase 1 Homo sapiens 45-49 22484158-7 2012 Interestingly, the profound up-regulation of HO-1 and Nrf-2 were observed in only ethanol-challenged cells, which evidenced that lucidone-induced induction of HO-/Nrf-2 were specific with oxidative stress. lucidone 129-137 heme oxygenase 1 Homo sapiens 45-49 22484158-8 2012 Thus, we concluded that lucidone-mediated up-regulation of phase-II enzymes and HO-1 via Nrf-2 signaling pathway may provide a pivotal mechanism for its hepatoprotective action. lucidone 24-32 heme oxygenase 1 Homo sapiens 80-84 22931644-1 2012 This study was aimed to investigate the effect of AMN107 (nilotinib) combined with heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin IX (ZnPPIX) on chronic myeloid leukemia (CML) cells and its mechanism. zinc protoporphyrin 117-139 heme oxygenase 1 Homo sapiens 101-105 22931644-1 2012 This study was aimed to investigate the effect of AMN107 (nilotinib) combined with heme oxygenase-1 (HO-1) inhibitor zinc protoporphyrin IX (ZnPPIX) on chronic myeloid leukemia (CML) cells and its mechanism. zinc protoporphyrin 141-147 heme oxygenase 1 Homo sapiens 101-105 22580386-1 2012 Antioxidant, anti-inflammatory and anti-atherogenic effects have been associated with elevations of unconjugated bilirubin (UCB) in serum and with the induction of heme oxygenase-1 (HO-1), the rate-limiting enzyme in UCB synthesis. ucb 217-220 heme oxygenase 1 Homo sapiens 164-180 22580386-1 2012 Antioxidant, anti-inflammatory and anti-atherogenic effects have been associated with elevations of unconjugated bilirubin (UCB) in serum and with the induction of heme oxygenase-1 (HO-1), the rate-limiting enzyme in UCB synthesis. ucb 217-220 heme oxygenase 1 Homo sapiens 182-186 22580386-4 2012 Moreover, induction of HO-1 with sodium arsenite led to 2.4-fold (p = 0.01) accumulation of intracellular UCB over basal level while sodium azide-derived oxidative stress resulted in a 60% drop (p < 0.001). sodium arsenite 33-48 heme oxygenase 1 Homo sapiens 23-27 22580386-4 2012 Moreover, induction of HO-1 with sodium arsenite led to 2.4-fold (p = 0.01) accumulation of intracellular UCB over basal level while sodium azide-derived oxidative stress resulted in a 60% drop (p < 0.001). Sodium Azide 133-145 heme oxygenase 1 Homo sapiens 23-27 22580386-7 2012 In conclusion, hyperbilirubinemia and HO-1 induction associated with inflammation and oxidative stress increase intracellular concentrations of UCB, thus enhancing the protection of cellular lipids against peroxidation. ucb 144-147 heme oxygenase 1 Homo sapiens 38-42 22580386-8 2012 Therefore, the previously reported protective effects of hyperbilirubinemia and HO-1 induction are at least in part due to intracellular accumulation of UCB. ucb 153-156 heme oxygenase 1 Homo sapiens 80-84 22753217-8 2012 Inhibition of HO activity by administration of tin mesoporphyrin to HF-fed mice transduced with the aP2-HO-1 reversed the decrease in Peg1/Mesoderm-specific transcript, TNFalpha, and MCP-1 levels. tin mesoporphyrin 47-64 heme oxygenase 1 Homo sapiens 104-108 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Heme 64-68 heme oxygenase 1 Homo sapiens 82-86 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 64-80 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 82-86 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Carbon Monoxide 156-171 heme oxygenase 1 Homo sapiens 64-80 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Carbon Monoxide 156-171 heme oxygenase 1 Homo sapiens 82-86 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Iron 181-185 heme oxygenase 1 Homo sapiens 64-80 22587389-4 2012 One of the key enzymes that control monocyte and DC function is haem oxygenase-1 (HO-1), which catalyses the degradation of the haem group into biliverdin, carbon monoxide and free iron. Iron 181-185 heme oxygenase 1 Homo sapiens 82-86 22484158-0 2012 Hepatoprotective effect of lucidone against alcohol-induced oxidative stress in human hepatic HepG2 cells through the up-regulation of HO-1/Nrf-2 antioxidant genes. lucidone 27-35 heme oxygenase 1 Homo sapiens 135-139 22484158-6 2012 Furthermore, Western blot and quantitative-PCR analyses showed that ethanol-exposure apparently down-regulated endogenous anti-oxidant hemoxygenase-1 (HO-1) expression, whereas pretreatment with lucidone significantly up-regulates HO-1 expression followed by the transcriptional activation of NF-E2 related factor-2 (Nrf-2). Ethanol 68-75 heme oxygenase 1 Homo sapiens 135-155 22484158-6 2012 Furthermore, Western blot and quantitative-PCR analyses showed that ethanol-exposure apparently down-regulated endogenous anti-oxidant hemoxygenase-1 (HO-1) expression, whereas pretreatment with lucidone significantly up-regulates HO-1 expression followed by the transcriptional activation of NF-E2 related factor-2 (Nrf-2). Ethanol 68-75 heme oxygenase 1 Homo sapiens 151-155 22484158-6 2012 Furthermore, Western blot and quantitative-PCR analyses showed that ethanol-exposure apparently down-regulated endogenous anti-oxidant hemoxygenase-1 (HO-1) expression, whereas pretreatment with lucidone significantly up-regulates HO-1 expression followed by the transcriptional activation of NF-E2 related factor-2 (Nrf-2). lucidone 195-203 heme oxygenase 1 Homo sapiens 151-155 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Heme 36-40 heme oxygenase 1 Homo sapiens 0-16 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Heme 36-40 heme oxygenase 1 Homo sapiens 18-22 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 0-16 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 18-22 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Biliverdine 63-73 heme oxygenase 1 Homo sapiens 0-16 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Biliverdine 63-73 heme oxygenase 1 Homo sapiens 18-22 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Iron 84-88 heme oxygenase 1 Homo sapiens 0-16 22302482-1 2012 Heme oxygenase-1 (HO-1) catabolizes heme into carbon monoxide, biliverdin, and free iron which mediate its protective effect against oxidative stress. Iron 84-88 heme oxygenase 1 Homo sapiens 18-22 22302482-6 2012 Treatment with the HO-1 inhibitor zinc protoporphyrin decreased the viability of colon cancer cell lines. zinc protoporphyrin 34-53 heme oxygenase 1 Homo sapiens 19-23 22593583-0 2012 Carbon monoxide mediates the anti-apoptotic effects of heme oxygenase-1 in medulloblastoma DAOY cells via K+ channel inhibition. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 55-71 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Heme 41-45 heme oxygenase 1 Homo sapiens 59-63 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Biliverdine 101-111 heme oxygenase 1 Homo sapiens 41-57 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Biliverdine 101-111 heme oxygenase 1 Homo sapiens 59-63 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). ammonium ferrous sulfate 113-119 heme oxygenase 1 Homo sapiens 41-57 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). ammonium ferrous sulfate 113-119 heme oxygenase 1 Homo sapiens 59-63 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 41-57 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 59-63 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 41-57 22593583-2 2012 Many cancer cells constitutively express heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 59-63 22579918-2 2012 Sustained induction of heme oxygenase-1 (HO-1) in nonerythroid cells plays a key role in many pathological processes, yet the effect of long-term HO-1 expression on cellular iron metabolism in the absence of exogenous heme is poorly understood. Heme 23-27 heme oxygenase 1 Homo sapiens 41-45 22579918-3 2012 Here we report that in a model nonerythroid cell, both transient and stable HO-1 expression increased heme oxygenase activity, but total cellular heme content was decreased only with transient enzyme expression. Heme 102-106 heme oxygenase 1 Homo sapiens 76-80 22579918-4 2012 Sustained HO-1 activity increased the expression of both the mitochondrial iron importer mitoferrin-2 and the rate-limiting enzyme in heme synthesis, aminolevulinate synthase-1, and it augmented the mitochondrial content of heme. Heme 134-138 heme oxygenase 1 Homo sapiens 10-14 22579918-4 2012 Sustained HO-1 activity increased the expression of both the mitochondrial iron importer mitoferrin-2 and the rate-limiting enzyme in heme synthesis, aminolevulinate synthase-1, and it augmented the mitochondrial content of heme. Heme 224-228 heme oxygenase 1 Homo sapiens 10-14 22579918-7 2012 Together, our results reveal a novel and coordinated adaptive response of nonerythroid cells to sustained HO-1 induction that has an impact on cellular iron homeostasis. Iron 152-156 heme oxygenase 1 Homo sapiens 106-110 22526619-7 2012 Furthermore, we found that oroxylin A treatment elevated intracellular reactive oxygen species levels and increased the protein expression level of two of the Nrf2 target genes heme oxygenase-1 and NADP(H):quinone oxidoreductase-1 in HCT-116 cells. 5,7-dihydroxy-6-methoxy-2-phenylchromen-4-one 27-37 heme oxygenase 1 Homo sapiens 177-193 22392462-0 2012 Curcumin ameliorates hydrogen peroxide-induced epithelial barrier disruption by upregulating heme oxygenase-1 expression in human intestinal epithelial cells. Curcumin 0-8 heme oxygenase 1 Homo sapiens 93-109 22139798-7 2012 HO-1 up-regulation in BCR-ABL1-expressing cells was suppressed by diphenyleneiodonium (DPI), a chemical inhibitor of the NADPH oxidase. diphenyleneiodonium 66-85 heme oxygenase 1 Homo sapiens 0-4 22139798-7 2012 HO-1 up-regulation in BCR-ABL1-expressing cells was suppressed by diphenyleneiodonium (DPI), a chemical inhibitor of the NADPH oxidase. diphenyleneiodonium 87-90 heme oxygenase 1 Homo sapiens 0-4 22392462-0 2012 Curcumin ameliorates hydrogen peroxide-induced epithelial barrier disruption by upregulating heme oxygenase-1 expression in human intestinal epithelial cells. Hydrogen Peroxide 21-38 heme oxygenase 1 Homo sapiens 93-109 22392462-2 2012 Heme oxygenase-1 (HO-1), which can be induced by curcumin (Cur), provides protection against various forms of oxidative stress. Curcumin 49-57 heme oxygenase 1 Homo sapiens 0-16 22392462-2 2012 Heme oxygenase-1 (HO-1), which can be induced by curcumin (Cur), provides protection against various forms of oxidative stress. Curcumin 49-57 heme oxygenase 1 Homo sapiens 18-22 22392462-10 2012 CONCLUSION: Cur protects human intestinal epithelial cells against H(2)O(2)-induced disruption of TJ and barrier dysfunction via the HO-1 pathway. Hydrogen Peroxide 67-75 heme oxygenase 1 Homo sapiens 133-137 22583702-1 2012 Heme oxygenase-1 (HMOX-1) is activated by oxidative stress, and gene responsiveness is reportedly determined by the number of dinucleotide (GT(n)) repeats in its highly polymorphic promoter region. Dinucleoside Phosphates 126-138 heme oxygenase 1 Homo sapiens 0-16 21129940-5 2012 Heme oxygenase-1, NAD(P)H: quinone oxidoreductase, glutathione-S-transferase, gamma-glutamyl cysteine ligase, and glutathione reductase are among the cytoprotective proteins induced by sulforaphane. sulforaphane 185-197 heme oxygenase 1 Homo sapiens 0-16 22583702-1 2012 Heme oxygenase-1 (HMOX-1) is activated by oxidative stress, and gene responsiveness is reportedly determined by the number of dinucleotide (GT(n)) repeats in its highly polymorphic promoter region. Dinucleoside Phosphates 126-138 heme oxygenase 1 Homo sapiens 18-24 22634503-6 2012 Arsenic-mediated ERK1/2 activation negatively regulated DNA polymerase beta expression and up regulated heme-oxygenase-1 at toxic concentrations. Arsenic 0-7 heme oxygenase 1 Homo sapiens 104-120 26105453-5 2012 HO-1 is also required to maintain nitric oxide synthesis (2). Nitric Oxide 34-46 heme oxygenase 1 Homo sapiens 0-4 22573121-2 2012 Calcitonin gene-related peptide (CGRP), which prevents circulating EPCs senescence and reverses Ang II-induced EPCs senescence is reduced in hypertensive patients, its level is stimulated by heme oxygenase-1 and is related with stimulation of nitric oxide. Nitric Oxide 243-255 heme oxygenase 1 Homo sapiens 191-207 22617388-0 2012 Bortezomib induces heme oxygenase-1 expression in multiple myeloma. Bortezomib 0-10 heme oxygenase 1 Homo sapiens 19-35 22617388-6 2012 Furthermore, we explored the expression of HO-1 in multiple myeloma cells in response to the key anti-myeloma drugs bortezomib and lenalidomide. Bortezomib 116-126 heme oxygenase 1 Homo sapiens 43-47 22617388-6 2012 Furthermore, we explored the expression of HO-1 in multiple myeloma cells in response to the key anti-myeloma drugs bortezomib and lenalidomide. Lenalidomide 131-143 heme oxygenase 1 Homo sapiens 43-47 22617388-7 2012 We show here for the first time that bortezomib increases HO-1 expression in a time- and concentration-dependent manner. Bortezomib 37-47 heme oxygenase 1 Homo sapiens 58-62 22617388-8 2012 Moreover, we also observe that HO-1 is increased in lenalidomide-resistant MM cell lines. Lenalidomide 52-64 heme oxygenase 1 Homo sapiens 31-35 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. reactive 202-210 heme oxygenase 1 Homo sapiens 21-60 22728708-10 2012 Interestingly, HO-1 expression was differently affected following treatment with various melatonin formulations. Melatonin 89-98 heme oxygenase 1 Homo sapiens 15-19 21352351-0 2012 Puerarin protects human umbilical vein endothelial cells against high glucose-induced apoptosis by upregulating heme oxygenase-1 and inhibiting calpain activation. puerarin 0-8 heme oxygenase 1 Homo sapiens 112-128 21352351-6 2012 HO-1 mRNA expression and HO activity were decreased in HUVECs treated with HG for 48 h. Compared with the group exposed to HG alone, co-incubation of HUVECs with puerarin and HG induced increases in HO-1 mRNA expression and HO activity. puerarin 162-170 heme oxygenase 1 Homo sapiens 0-4 21352351-6 2012 HO-1 mRNA expression and HO activity were decreased in HUVECs treated with HG for 48 h. Compared with the group exposed to HG alone, co-incubation of HUVECs with puerarin and HG induced increases in HO-1 mRNA expression and HO activity. puerarin 162-170 heme oxygenase 1 Homo sapiens 199-203 21352351-7 2012 The HO-1 inhibitor protoporphyrin IX zinc (II) abolished the inhibitory effect of puerarin on HG-induced calpain and caspase-3 activation, as well as apoptosis. protoporphyrin IX 19-36 heme oxygenase 1 Homo sapiens 4-8 21352351-7 2012 The HO-1 inhibitor protoporphyrin IX zinc (II) abolished the inhibitory effect of puerarin on HG-induced calpain and caspase-3 activation, as well as apoptosis. puerarin 82-90 heme oxygenase 1 Homo sapiens 4-8 21352351-8 2012 The data show that puerarin protects against HG-induced endothelial cell apoptosis by a mechanism involving upregulation of HO-1 expression and inhibition of calpain activity. puerarin 19-27 heme oxygenase 1 Homo sapiens 124-128 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. reactive 202-210 heme oxygenase 1 Homo sapiens 62-66 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. oxygen species 211-225 heme oxygenase 1 Homo sapiens 21-60 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. oxygen species 211-225 heme oxygenase 1 Homo sapiens 62-66 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. Nitrogen 239-247 heme oxygenase 1 Homo sapiens 21-60 22549002-3 2012 The induction of the heme oxygenase-1/biliverdin reductase-A (HO-1/BVR-A) system in the brain represents one of the earliest mechanisms activated by cells to counteract the noxious effects of increased reactive oxygen species and reactive nitrogen species. Nitrogen 239-247 heme oxygenase 1 Homo sapiens 62-66 22549002-10 2012 Interestingly, despite the lack of oxidative stress-induced AD neuropathology in cerebellum, HO-1 demonstrated increased Ser-residue phosphorylation and oxidative posttranslational modifications in this brain area, suggesting HO-1 as a target of oxidative damage even in the cerebellum. Serine 121-124 heme oxygenase 1 Homo sapiens 93-97 22943759-1 2012 OBJECTIVE: To investigate whether chlorophyllin could protect human umbilical vein endothelial cell (HUVEC) against oxidative damage by inducing the expression of heme oxygenase-1 (HO-1) and to explore the underlying mechanism. chlorophyllin 34-47 heme oxygenase 1 Homo sapiens 163-179 22447044-0 2012 Protein kinase CK2 mediates peroxynitrite-induced heme oxygenase-1 expression in articular chondrocytes. Peroxynitrous Acid 28-41 heme oxygenase 1 Homo sapiens 50-66 22447044-2 2012 In the present study, we examined the role of protein kinase casein kinase (CK2) on peroxynitrite-induced expression of HO-1 in primary articular chondrocytes. Peroxynitrous Acid 96-109 heme oxygenase 1 Homo sapiens 132-136 22271370-2 2012 Heme oxygenase (HO)-1, a rate-limiting enzyme of heme degradation, plays a protective role against oxidative stress. Heme 49-53 heme oxygenase 1 Homo sapiens 0-21 22447044-11 2012 Taken together, our findings suggest that peroxynitrite activates Nrf2 via CK2 signaling, leading to the upregulation of HO-1 in primary chondrocytes. Peroxynitrous Acid 42-55 heme oxygenase 1 Homo sapiens 133-137 22440905-8 2012 In iron-challenged, cultured proximal tubule cells, we observed a positive correlation between HO-1 mRNA level and HO-1 release. Iron 3-7 heme oxygenase 1 Homo sapiens 95-99 22440905-8 2012 In iron-challenged, cultured proximal tubule cells, we observed a positive correlation between HO-1 mRNA level and HO-1 release. Iron 3-7 heme oxygenase 1 Homo sapiens 115-119 22394342-6 2012 Due to the cumulative effects of HO-1 on heme catabolism and the generation of biologically active downstream products, induction of HO-1 might serve as a protective mechanism against oxidative stress and inflammation-induced injury. Heme 41-45 heme oxygenase 1 Homo sapiens 33-37 22394342-6 2012 Due to the cumulative effects of HO-1 on heme catabolism and the generation of biologically active downstream products, induction of HO-1 might serve as a protective mechanism against oxidative stress and inflammation-induced injury. Heme 41-45 heme oxygenase 1 Homo sapiens 133-137 22394342-8 2012 This review highlights the roles of HO-1 in lung disease induced by environmental toxins such as cigarette smoke (CS), silica, and asbestos. Cesium 114-116 heme oxygenase 1 Homo sapiens 36-40 22394342-8 2012 This review highlights the roles of HO-1 in lung disease induced by environmental toxins such as cigarette smoke (CS), silica, and asbestos. Silicon Dioxide 119-125 heme oxygenase 1 Homo sapiens 36-40 22943759-1 2012 OBJECTIVE: To investigate whether chlorophyllin could protect human umbilical vein endothelial cell (HUVEC) against oxidative damage by inducing the expression of heme oxygenase-1 (HO-1) and to explore the underlying mechanism. chlorophyllin 34-47 heme oxygenase 1 Homo sapiens 181-185 22943759-7 2012 Chlorophyllin treatment could induce expression of HO-1 in a dose- and time-dependent manner. chlorophyllin 0-13 heme oxygenase 1 Homo sapiens 51-55 22943759-9 2012 LY294002, the specific inhibitor of PI3K, could suppress the activation of PI3K/Akt and the induced expression of HO-1 in a dose-dependent manner. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 0-8 heme oxygenase 1 Homo sapiens 114-118 22943759-11 2012 Up-regulation of HO-1 expression plays a pivotal role in the protection of chlorophyllin, while the activation of PI3K/Akt signaling pathway is required in the induction of HO-1. chlorophyllin 75-88 heme oxygenase 1 Homo sapiens 17-21 22155346-7 2012 ATO activated a silent Nrf2 pathway in cultured OSCC cells as shown by induction of Nrf2 and Nrf2-regulated genes, NAD(P)H:quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), in a dose-dependent manner. Arsenic Trioxide 0-3 heme oxygenase 1 Homo sapiens 159-175 22586396-7 2012 Heme is degraded by heme oxygenase (HO) that exists as two isoforms: inducible HO-1 and constitutively expressed HO-2. Heme 0-4 heme oxygenase 1 Homo sapiens 79-83 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Heme 32-36 heme oxygenase 1 Homo sapiens 0-16 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Heme 32-36 heme oxygenase 1 Homo sapiens 18-21 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Biliverdine 40-50 heme oxygenase 1 Homo sapiens 0-16 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Biliverdine 40-50 heme oxygenase 1 Homo sapiens 18-21 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Carbon Monoxide 52-67 heme oxygenase 1 Homo sapiens 0-16 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Carbon Monoxide 52-67 heme oxygenase 1 Homo sapiens 18-21 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Iron 77-81 heme oxygenase 1 Homo sapiens 0-16 22503972-4 2012 Heme oxygenase 1 (HO1) degrades heme to biliverdin, carbon monoxide and free iron, and is a stress-responsive protein. Iron 77-81 heme oxygenase 1 Homo sapiens 18-21 22503972-10 2012 Taken together, our findings point to three novel functions of STC2, and suggest that STC2 interacts with HO1 to form a eukaryotic "stressosome" involved in the degradation of heme. Heme 176-180 heme oxygenase 1 Homo sapiens 106-109 22426130-8 2012 siRNA-mediated silencing of VEGFA or VEGFR2 could reverse the inhibitory effect of HO-1 on VSMC migration. vsmc 91-95 heme oxygenase 1 Homo sapiens 83-87 22386815-0 2012 Ellagic acid protects human keratinocyte (HaCaT) cells against UVA-induced oxidative stress and apoptosis through the upregulation of the HO-1 and Nrf-2 antioxidant genes. Ellagic Acid 0-12 heme oxygenase 1 Homo sapiens 138-142 22431362-3 2012 The introduction of an oxygen atom in the alkyl linker produced analogues with decreased potency toward HO-1, whereas the presence of a sulfur atom in the linker gave rise to analogues with greater potency toward HO-1 than the carbon-containing analogues. Sulfur 136-142 heme oxygenase 1 Homo sapiens 213-217 22386815-7 2012 Notably, the antioxidant potential of ellagic acid was directly correlated with the increased expression of HO-1 and SOD, which was followed by the downregulation of Keap1 and the augmented nuclear translocation and transcriptional activation of Nrf2 with or without UVA irradiation. Ellagic Acid 38-50 heme oxygenase 1 Homo sapiens 108-120 22201606-2 2012 The inducible stress protein heme oxygenase-1 (HO-1) has been implicated in cytoprotection against the toxic action of many xenobiotics, including CS. Cesium 147-149 heme oxygenase 1 Homo sapiens 29-45 22201606-2 2012 The inducible stress protein heme oxygenase-1 (HO-1) has been implicated in cytoprotection against the toxic action of many xenobiotics, including CS. Cesium 147-149 heme oxygenase 1 Homo sapiens 47-51 22201606-6 2012 Carbon monoxide (CO), a primary reaction product of HO-1 has been implicated in cytoprotection in many acute lung injury models, though it"s precise role in chronic CS-induced lung injury remains unclear. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 52-56 22201606-6 2012 Carbon monoxide (CO), a primary reaction product of HO-1 has been implicated in cytoprotection in many acute lung injury models, though it"s precise role in chronic CS-induced lung injury remains unclear. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 52-56 22201606-9 2012 The implications of the cytoprotective potential of HO-1/CO system in CS-induced lung injury and COPD are discussed. Cesium 70-72 heme oxygenase 1 Homo sapiens 52-56 22305882-6 2012 Furthermore, the cytoprotective enzyme heme oxygenase-1 could be upregulated in leonurine-treated HUVEC. leonurine 80-89 heme oxygenase 1 Homo sapiens 39-55 22326969-6 2012 Instead, the over expression of anti-apoptotic Bcl-2, anti-oxidative heme oxygenase-1 (HO-1) and cell cycle regulator p16INK4 seemed to be more important for the gaining of cisplatin in these human urothelial carcinoma cell. Cisplatin 173-182 heme oxygenase 1 Homo sapiens 69-85 22781797-0 2012 [The effect of retrovirus-mediated HO-1 gene on chronic myeloid leukemia resistance cell K562/A02 apoptosis induced by nilotinib]. nilotinib 119-128 heme oxygenase 1 Homo sapiens 35-39 22781797-1 2012 OBJECTIVE: To investigate the effect of retrovirus mediated heme oxygenase (HO)-1 gene on chronic myeloid leukemia (CML) resistance cell apoptosis induced by nilotinib (AMN107). nilotinib 158-167 heme oxygenase 1 Homo sapiens 60-81 22421218-0 2012 Smad7 sensitizes A549 lung cancer cells to cisplatin-induced apoptosis through heme oxygenase-1 inhibition. Cisplatin 43-52 heme oxygenase 1 Homo sapiens 79-95 22421218-4 2012 The HO-1 protein level was elevated in cisplatin-resistant A549 human lung cancer cells and blockade of HO-1 activation sensitized the cells to apoptosis. Cisplatin 39-48 heme oxygenase 1 Homo sapiens 4-8 22421218-4 2012 The HO-1 protein level was elevated in cisplatin-resistant A549 human lung cancer cells and blockade of HO-1 activation sensitized the cells to apoptosis. Cisplatin 39-48 heme oxygenase 1 Homo sapiens 104-108 22421218-7 2012 Consistently, Smad7 sensitized A549 cells to cisplatin-induced apoptosis and these effects were dependent on HO-1 and Akt inhibition. Cisplatin 45-54 heme oxygenase 1 Homo sapiens 109-113 22492492-0 2012 Concomitant induction of heme oxygenase-1 attenuates the cytotoxicity of arsenic species from lumbricus extract in human liver HepG2 cells. Arsenic 73-80 heme oxygenase 1 Homo sapiens 25-41 22147556-2 2012 The production of nitric oxide by the conversion of l-(2,3,4,5)-[3H] arginine to l-(3H) citrulline, the activity of haem oxygenase 1 (HO-1) through bilirubin synthesis and the expression of inducible nitric oxide synthase (iNOS), HO-1 proteins and messenger RNA by Western blot and reverse-transcribed polymerase chain reaction were also tested. Nitric Oxide 18-30 heme oxygenase 1 Homo sapiens 116-132 22147556-2 2012 The production of nitric oxide by the conversion of l-(2,3,4,5)-[3H] arginine to l-(3H) citrulline, the activity of haem oxygenase 1 (HO-1) through bilirubin synthesis and the expression of inducible nitric oxide synthase (iNOS), HO-1 proteins and messenger RNA by Western blot and reverse-transcribed polymerase chain reaction were also tested. Nitric Oxide 18-30 heme oxygenase 1 Homo sapiens 134-138 22147556-2 2012 The production of nitric oxide by the conversion of l-(2,3,4,5)-[3H] arginine to l-(3H) citrulline, the activity of haem oxygenase 1 (HO-1) through bilirubin synthesis and the expression of inducible nitric oxide synthase (iNOS), HO-1 proteins and messenger RNA by Western blot and reverse-transcribed polymerase chain reaction were also tested. Nitric Oxide 18-30 heme oxygenase 1 Homo sapiens 230-234 22147556-2 2012 The production of nitric oxide by the conversion of l-(2,3,4,5)-[3H] arginine to l-(3H) citrulline, the activity of haem oxygenase 1 (HO-1) through bilirubin synthesis and the expression of inducible nitric oxide synthase (iNOS), HO-1 proteins and messenger RNA by Western blot and reverse-transcribed polymerase chain reaction were also tested. Bilirubin 148-157 heme oxygenase 1 Homo sapiens 116-132 22147556-2 2012 The production of nitric oxide by the conversion of l-(2,3,4,5)-[3H] arginine to l-(3H) citrulline, the activity of haem oxygenase 1 (HO-1) through bilirubin synthesis and the expression of inducible nitric oxide synthase (iNOS), HO-1 proteins and messenger RNA by Western blot and reverse-transcribed polymerase chain reaction were also tested. Bilirubin 148-157 heme oxygenase 1 Homo sapiens 134-138 22147556-5 2012 The results revealed an over-expression of iNOS and HO-1 in the papilla, compared with that in the pulp, mediated by the nuclear factor kappa B transcription factor activated by the reactive oxygen species that acts as scavengers for the superoxide radicals. Reactive Oxygen Species 182-205 heme oxygenase 1 Homo sapiens 52-56 22147556-5 2012 The results revealed an over-expression of iNOS and HO-1 in the papilla, compared with that in the pulp, mediated by the nuclear factor kappa B transcription factor activated by the reactive oxygen species that acts as scavengers for the superoxide radicals. Superoxides 238-248 heme oxygenase 1 Homo sapiens 52-56 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . protoporphyrin IX 103-120 heme oxygenase 1 Homo sapiens 84-88 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . S-Nitroso-N-propionyl-D,L-penicillamine 122-126 heme oxygenase 1 Homo sapiens 84-88 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 162-170 heme oxygenase 1 Homo sapiens 186-190 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Heme 73-77 heme oxygenase 1 Homo sapiens 0-16 22492492-9 2012 These results suggest that arsenic-containing compounds are responsible for HO-1 induction by Lumbricus extract. Arsenic 27-34 heme oxygenase 1 Homo sapiens 76-80 22492492-10 2012 Although the exact role of toxic arsenic compounds in the treatment of oxidative injury remains unclear, concomitant HO-1 induction may be a key mechanism to antagonize the cytotoxicity of arsenic compounds in human cells. Arsenic 189-196 heme oxygenase 1 Homo sapiens 117-121 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Heme 73-77 heme oxygenase 1 Homo sapiens 18-22 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Biliverdine 97-107 heme oxygenase 1 Homo sapiens 0-16 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Biliverdine 97-107 heme oxygenase 1 Homo sapiens 18-22 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 0-16 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 18-22 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Carbon Monoxide 126-128 heme oxygenase 1 Homo sapiens 0-16 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Carbon Monoxide 126-128 heme oxygenase 1 Homo sapiens 18-22 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Iron 135-139 heme oxygenase 1 Homo sapiens 0-16 22492492-1 2012 Heme oxygenase-1 (HO-1) is an inducible antioxidant enzyme that degrades heme to three products, biliverdin, carbon monoxide (CO), and iron ion. Iron 135-139 heme oxygenase 1 Homo sapiens 18-22 22492492-7 2012 Both p38 MAP kinase and NF-E2-related factor 2 (Nrf2) pathways were found to modulate HO-1 induction by Lumbricus extract and NaAsO(2) . naaso 126-131 heme oxygenase 1 Homo sapiens 86-90 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . arsenite 20-28 heme oxygenase 1 Homo sapiens 84-88 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . arsenite 20-28 heme oxygenase 1 Homo sapiens 186-190 22492492-8 2012 The cytotoxicity of arsenite was augmented by p38 MAP kinase inhibitor SB202190 and HO-1 inhibitor tin protoporphyrin IX (SnPP), whereas p38 MAP kinase inhibitor SB202190 also inhibited HO-1 induction by NaAsO(2) . Tin 99-102 heme oxygenase 1 Homo sapiens 84-88 22490514-8 2012 Whether the induction or inhibition of HO-1 by cobalt-protoporphyrin-IX (CoPP) or zinc-protoporphyrin-IX (ZnPP) could affect the sensitivity of MKN-45 cells to cisplatin was also studied. cobaltiprotoporphyrin 47-71 heme oxygenase 1 Homo sapiens 39-43 22690236-8 2012 HO-1 mRNA expression was positively correlated with levels of carboxyhemoglobin, serum ferritin, and serum MDA and negatively correlated with levels of erythrocyte GSH in CLD patients. Glutathione 164-167 heme oxygenase 1 Homo sapiens 0-4 22490514-8 2012 Whether the induction or inhibition of HO-1 by cobalt-protoporphyrin-IX (CoPP) or zinc-protoporphyrin-IX (ZnPP) could affect the sensitivity of MKN-45 cells to cisplatin was also studied. cobaltiprotoporphyrin 73-77 heme oxygenase 1 Homo sapiens 39-43 22490514-8 2012 Whether the induction or inhibition of HO-1 by cobalt-protoporphyrin-IX (CoPP) or zinc-protoporphyrin-IX (ZnPP) could affect the sensitivity of MKN-45 cells to cisplatin was also studied. zinc protoporphyrin 106-110 heme oxygenase 1 Homo sapiens 39-43 22490514-8 2012 Whether the induction or inhibition of HO-1 by cobalt-protoporphyrin-IX (CoPP) or zinc-protoporphyrin-IX (ZnPP) could affect the sensitivity of MKN-45 cells to cisplatin was also studied. Cisplatin 160-169 heme oxygenase 1 Homo sapiens 39-43 22490514-10 2012 HO-1 overexpression could lead to an increased resistance to cisplatin, whereas down-regulation of HO-1 expression by siRNA or chemical inhibition of HO-1 could lead to increased chemosensitivity to cisplatin in MKN-45 cells. Cisplatin 61-70 heme oxygenase 1 Homo sapiens 0-4 22490514-10 2012 HO-1 overexpression could lead to an increased resistance to cisplatin, whereas down-regulation of HO-1 expression by siRNA or chemical inhibition of HO-1 could lead to increased chemosensitivity to cisplatin in MKN-45 cells. Cisplatin 199-208 heme oxygenase 1 Homo sapiens 99-103 22490514-10 2012 HO-1 overexpression could lead to an increased resistance to cisplatin, whereas down-regulation of HO-1 expression by siRNA or chemical inhibition of HO-1 could lead to increased chemosensitivity to cisplatin in MKN-45 cells. Cisplatin 199-208 heme oxygenase 1 Homo sapiens 99-103 22197494-8 2012 Resveratrol prevented the up-regulation of early growth response protein-1 (Egr-1), a transcription factor necessary for induction of the vascular endothelial growth factor receptor-1 gene and caused up-regulation of heme oxygenase-1, a cytoprotective enzyme found to be dysfunctional in preeclampsia. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 217-233 22098251-8 2012 RESULTS: Ex vivo, donor liver preconditioning with Nodosin perfusion induced HO-1 expression and enzyme activity significantly, compared with the control group (P < 0.05). nodosin 51-58 heme oxygenase 1 Homo sapiens 77-81 22240980-8 2012 Reactive oxygen species (ROS) were detected along with the formation of protein carbonyls and the induction of heme oxygenase-1. Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 111-127 22240980-8 2012 Reactive oxygen species (ROS) were detected along with the formation of protein carbonyls and the induction of heme oxygenase-1. Reactive Oxygen Species 25-28 heme oxygenase 1 Homo sapiens 111-127 21971692-6 2012 It was found that EEPV increased HO-1 protein expression in RAW 264.7 cells, which was significantly inhibited by LY294002, but not PD98059, SB203580 or SP600125. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 114-122 heme oxygenase 1 Homo sapiens 33-37 21971692-7 2012 In addition, EEPV activated NF-E2-related factor (Nrf2) to move from the cytosol to the nucleus, and EEPV-induced HO-1 and activation of ARE-luciferase activity were significantly reduced by siNrf2 transfection and LY294002 but not SB203508. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 215-223 heme oxygenase 1 Homo sapiens 114-118 21971692-7 2012 In addition, EEPV activated NF-E2-related factor (Nrf2) to move from the cytosol to the nucleus, and EEPV-induced HO-1 and activation of ARE-luciferase activity were significantly reduced by siNrf2 transfection and LY294002 but not SB203508. sb203508 232-240 heme oxygenase 1 Homo sapiens 114-118 22197494-9 2012 CONCLUSION: In summary, resveratrol can inhibit sFlt-1 release and up-regulate heme oxygenase-1; thus, may offer therapeutic potential in preeclampsia. Resveratrol 24-35 heme oxygenase 1 Homo sapiens 79-95 22166397-2 2012 In this study, we examined whether M. cordata extract and/or its major alkaloid constituents, protopine, allocryptopine, sanguinarine and chelerythrine activate the Nrf2 signalling pathway which regulates the expression of cytoprotective enzymes including heme oxygenase-1 (HO-1) and thioredoxin 1. chelerythrine 138-151 heme oxygenase 1 Homo sapiens 274-278 21954917-4 2012 Treatment with the HO-1 inducer CoPP (cobalt protoporphyrin IX) counteracted the stimulatory effects of IL-1beta on IL-6, nitrite, PGE2 (prostaglandin E2), TGF (transforming growth factor) beta2, TGFbeta3 and osteocalcin. cobaltiprotoporphyrin 38-62 heme oxygenase 1 Homo sapiens 19-23 21954917-4 2012 Treatment with the HO-1 inducer CoPP (cobalt protoporphyrin IX) counteracted the stimulatory effects of IL-1beta on IL-6, nitrite, PGE2 (prostaglandin E2), TGF (transforming growth factor) beta2, TGFbeta3 and osteocalcin. Nitrites 122-129 heme oxygenase 1 Homo sapiens 19-23 21954917-4 2012 Treatment with the HO-1 inducer CoPP (cobalt protoporphyrin IX) counteracted the stimulatory effects of IL-1beta on IL-6, nitrite, PGE2 (prostaglandin E2), TGF (transforming growth factor) beta2, TGFbeta3 and osteocalcin. Dinoprostone 131-135 heme oxygenase 1 Homo sapiens 19-23 21954917-4 2012 Treatment with the HO-1 inducer CoPP (cobalt protoporphyrin IX) counteracted the stimulatory effects of IL-1beta on IL-6, nitrite, PGE2 (prostaglandin E2), TGF (transforming growth factor) beta2, TGFbeta3 and osteocalcin. Dinoprostone 137-153 heme oxygenase 1 Homo sapiens 19-23 22166397-8 2012 Nonetheless, sanguinarine at micromolar, non-cytotoxic concentrations elevated protein levels of HO-1 and thioredoxin 1 in primary cultures of human hepatocytes. sanguinarine 13-25 heme oxygenase 1 Homo sapiens 97-101 22203369-1 2012 Probucol inhibits the proliferation of vascular smooth muscle cells in vitro and in vivo, and the drug reduces intimal hyperplasia and atherosclerosis in animals via induction of heme oxygenase-1 (HO-1). Probucol 0-8 heme oxygenase 1 Homo sapiens 179-195 22203369-1 2012 Probucol inhibits the proliferation of vascular smooth muscle cells in vitro and in vivo, and the drug reduces intimal hyperplasia and atherosclerosis in animals via induction of heme oxygenase-1 (HO-1). Probucol 0-8 heme oxygenase 1 Homo sapiens 197-201 22203369-4 2012 However, whereas inhibition of HO-1 reversed the antiproliferative effects of probucol, this was not observed with succinobucol. Probucol 78-86 heme oxygenase 1 Homo sapiens 31-35 21725851-1 2012 Heme oxygenase-1 (HO-1) which is a rate-limiting enzyme in heme degradation processes shows a dinucleotide GT repeat in the promoter that alters the level of gene transcription. Heme 59-63 heme oxygenase 1 Homo sapiens 0-16 21647550-2 2012 On the other hand, oxidative stress has been implicated in the pathogenesis of age-related macular degeneration (AMD) and heme oxygenase-1 (HO-1), encoded by the HMOX1 gene and heme oxygenase-2 (HO-2), encoded by the HMOX2 gene are important markers of iron-related oxidative stress and its consequences. Iron 253-257 heme oxygenase 1 Homo sapiens 162-167 21725851-1 2012 Heme oxygenase-1 (HO-1) which is a rate-limiting enzyme in heme degradation processes shows a dinucleotide GT repeat in the promoter that alters the level of gene transcription. Heme 59-63 heme oxygenase 1 Homo sapiens 18-22 21647550-2 2012 On the other hand, oxidative stress has been implicated in the pathogenesis of age-related macular degeneration (AMD) and heme oxygenase-1 (HO-1), encoded by the HMOX1 gene and heme oxygenase-2 (HO-2), encoded by the HMOX2 gene are important markers of iron-related oxidative stress and its consequences. Iron 253-257 heme oxygenase 1 Homo sapiens 122-138 21725851-1 2012 Heme oxygenase-1 (HO-1) which is a rate-limiting enzyme in heme degradation processes shows a dinucleotide GT repeat in the promoter that alters the level of gene transcription. Dinucleoside Phosphates 94-106 heme oxygenase 1 Homo sapiens 0-16 21647550-2 2012 On the other hand, oxidative stress has been implicated in the pathogenesis of age-related macular degeneration (AMD) and heme oxygenase-1 (HO-1), encoded by the HMOX1 gene and heme oxygenase-2 (HO-2), encoded by the HMOX2 gene are important markers of iron-related oxidative stress and its consequences. Iron 253-257 heme oxygenase 1 Homo sapiens 140-144 21725851-1 2012 Heme oxygenase-1 (HO-1) which is a rate-limiting enzyme in heme degradation processes shows a dinucleotide GT repeat in the promoter that alters the level of gene transcription. Dinucleoside Phosphates 94-106 heme oxygenase 1 Homo sapiens 18-22 22200816-0 2012 Sulforaphane protects against 6-hydroxydopamine-induced cytotoxicity by increasing expression of heme oxygenase-1 in a PI3K/Akt-dependent manner. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 109-125 22200816-0 2012 Sulforaphane protects against 6-hydroxydopamine-induced cytotoxicity by increasing expression of heme oxygenase-1 in a PI3K/Akt-dependent manner. Oxidopamine 30-47 heme oxygenase 1 Homo sapiens 109-125 22200816-7 2012 SF also increased heme oxygenase-1 (HO-1) expression, which conferred protection against 6-OHDA-induced cytotoxicity. Oxidopamine 113-119 heme oxygenase 1 Homo sapiens 30-46 22200816-7 2012 SF also increased heme oxygenase-1 (HO-1) expression, which conferred protection against 6-OHDA-induced cytotoxicity. Oxidopamine 113-119 heme oxygenase 1 Homo sapiens 48-52 22200816-9 2012 These results suggest that SF inhibits 6-OHDA-induced cytotoxicity through increasing HO-1 expression in a PI3K/Akt-dependent manner. Oxidopamine 51-57 heme oxygenase 1 Homo sapiens 98-102 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Heme 102-106 heme oxygenase 1 Homo sapiens 0-16 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Heme 102-106 heme oxygenase 1 Homo sapiens 18-22 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Carbon Monoxide 112-127 heme oxygenase 1 Homo sapiens 0-16 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Carbon Monoxide 112-127 heme oxygenase 1 Homo sapiens 18-22 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Bilirubin 142-151 heme oxygenase 1 Homo sapiens 0-16 22200625-5 2012 Heme oxygenase-1 (HO-1) is the inducible isoform of the first and rate-limiting enzyme which degrades heme into carbon monoxide, ferritin and bilirubin. Bilirubin 142-151 heme oxygenase 1 Homo sapiens 18-22 25774181-2 2012 Graded concentration and time course experiments demonstrate that curcumin significantly upregulates phosphatidylinositol 3-kinase (PI3K), Akt, nuclear factor E2-related factor-2 (Nrf2), heme oxygenase 1 and ferritin expression, and that it significantly downregulates heme oxygenase 2, reactive oxygen species and amyloid-beta 40/42 expression. Curcumin 66-74 heme oxygenase 1 Homo sapiens 187-203 22100389-0 2012 Dexamethasone-loaded peptide micelles for delivery of the heme oxygenase-1 gene to ischemic brain. Dexamethasone 0-13 heme oxygenase 1 Homo sapiens 58-74 25774181-4 2012 The results indicate that the cytoprotection conferred by curcumin on APPswe transfected SH-SY5Y cells is mediated by its ability to regulate the balance between heme oxygenase 1 and 2 via the PI3K/Akt/Nrf2 intracellular signaling pathway. Curcumin 58-66 heme oxygenase 1 Homo sapiens 162-184 21997482-10 2012 1,2-NQ exposure also causes marked expression of HO-1 that appears to be enhanced by suppression of H2O2. 1,2-naphthoquinone 0-6 heme oxygenase 1 Homo sapiens 49-53 22260728-0 2012 Apo-8"-lycopenal induces expression of HO-1 and NQO-1 via the ERK/p38-Nrf2-ARE pathway in human HepG2 cells. apo-8'-lycopenal 0-16 heme oxygenase 1 Homo sapiens 39-43 22155307-5 2012 HO-1 was induced with cobalt protoporphyrin IX (CoPP) and by transduction with LV-HO-1. cobaltiprotoporphyrin 22-46 heme oxygenase 1 Homo sapiens 0-4 22155307-5 2012 HO-1 was induced with cobalt protoporphyrin IX (CoPP) and by transduction with LV-HO-1. cobaltiprotoporphyrin 48-52 heme oxygenase 1 Homo sapiens 0-4 22155307-8 2012 HO-1 also exerted inhibitory effects on prostaglandin (PG)E(2) production which could be dependent on cyclooxygenase-2 and microsomal PGE synthase-1 down-regulation. Dinoprostone 40-62 heme oxygenase 1 Homo sapiens 0-4 22095074-0 2012 Guggulsterone induces heme oxygenase-1 expression through activation of Nrf2 in human mammary epithelial cells: PTEN as a putative target. pregna-4,17-diene-3,16-dione 0-13 heme oxygenase 1 Homo sapiens 22-38 22095074-11 2012 Pretreatment with the thiol-reducing agents attenuated Akt phosphorylation, Nrf2 activation and HO-1 expression, suggesting that cis-GS may cause thiol modification of an upstream signaling modulator. Sulfhydryl Compounds 22-27 heme oxygenase 1 Homo sapiens 96-100 22095074-11 2012 Pretreatment with the thiol-reducing agents attenuated Akt phosphorylation, Nrf2 activation and HO-1 expression, suggesting that cis-GS may cause thiol modification of an upstream signaling modulator. pregna-4,17-diene-3,16-dione 129-135 heme oxygenase 1 Homo sapiens 96-100 22095074-13 2012 The mutation in cysteine 124 present in the catalytic domain of PTEN abolished cis-GS-induced HO-1 expression as well as Akt phosphorylation. Cysteine 16-24 heme oxygenase 1 Homo sapiens 94-98 21992109-1 2012 HO-1 (haem oxygenase-1) is a ubiquitously expressed inducible enzyme degrading haem to CO, biliverdin and Fe(2+). Carbon Monoxide 87-89 heme oxygenase 1 Homo sapiens 0-22 21992109-1 2012 HO-1 (haem oxygenase-1) is a ubiquitously expressed inducible enzyme degrading haem to CO, biliverdin and Fe(2+). Biliverdine 91-101 heme oxygenase 1 Homo sapiens 0-22 21992109-1 2012 HO-1 (haem oxygenase-1) is a ubiquitously expressed inducible enzyme degrading haem to CO, biliverdin and Fe(2+). ammonium ferrous sulfate 106-112 heme oxygenase 1 Homo sapiens 0-22 21997482-10 2012 1,2-NQ exposure also causes marked expression of HO-1 that appears to be enhanced by suppression of H2O2. Hydrogen Peroxide 100-104 heme oxygenase 1 Homo sapiens 49-53 22019695-0 2012 Reactive oxygen species and PI3K/Akt signaling play key roles in the induction of Nrf2-driven heme oxygenase-1 expression in sulforaphane-treated human mesothelioma MSTO-211H cells. Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 94-110 22019695-0 2012 Reactive oxygen species and PI3K/Akt signaling play key roles in the induction of Nrf2-driven heme oxygenase-1 expression in sulforaphane-treated human mesothelioma MSTO-211H cells. sulforaphane 125-137 heme oxygenase 1 Homo sapiens 94-110 22155056-1 2012 Sulforaphane (SFN) is a dietary isothiocyanate that exerts chemopreventive effects via NF-E2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). sulforaphane 14-17 heme oxygenase 1 Homo sapiens 177-193 22019695-2 2012 In this study, we investigated Nrf2/HO-1 induction in response to sulforaphane and determined the signaling pathways involved in this process. sulforaphane 66-78 heme oxygenase 1 Homo sapiens 36-40 22019695-7 2012 Activation of the Nrf2/HO-1 system after sulforaphane treatment was suppressed by pretreatment with NAC or Ly294002, a PI3K inhibitor. sulforaphane 41-53 heme oxygenase 1 Homo sapiens 23-27 22019695-7 2012 Activation of the Nrf2/HO-1 system after sulforaphane treatment was suppressed by pretreatment with NAC or Ly294002, a PI3K inhibitor. Acetylcysteine 100-103 heme oxygenase 1 Homo sapiens 23-27 22019695-7 2012 Activation of the Nrf2/HO-1 system after sulforaphane treatment was suppressed by pretreatment with NAC or Ly294002, a PI3K inhibitor. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 107-115 heme oxygenase 1 Homo sapiens 23-27 22019695-8 2012 Knockdown of Nrf2 with siRNA decreased cell viability and attenuated sulforaphane-induced HO-1 up-regulation. sulforaphane 69-81 heme oxygenase 1 Homo sapiens 90-94 22019695-9 2012 Overall, our results indicate that ROS generation and/or activation of PI3K/Akt signaling regulate cell survival and Nrf2-driven HO-1 expression in sulforaphane-treated MSTO-211H cells. Reactive Oxygen Species 35-38 heme oxygenase 1 Homo sapiens 129-133 22019695-9 2012 Overall, our results indicate that ROS generation and/or activation of PI3K/Akt signaling regulate cell survival and Nrf2-driven HO-1 expression in sulforaphane-treated MSTO-211H cells. sulforaphane 148-160 heme oxygenase 1 Homo sapiens 129-133 22155056-1 2012 Sulforaphane (SFN) is a dietary isothiocyanate that exerts chemopreventive effects via NF-E2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). sulforaphane 0-12 heme oxygenase 1 Homo sapiens 177-193 22155056-1 2012 Sulforaphane (SFN) is a dietary isothiocyanate that exerts chemopreventive effects via NF-E2-related factor 2 (Nrf2)-mediated induction of antioxidant/phase II enzymes, such as heme oxygenase-1 (HO-1) and NAD(P)H quinone oxidoreductase 1 (NQO1). isothiocyanic acid 32-46 heme oxygenase 1 Homo sapiens 177-193 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Iron 114-118 heme oxygenase 1 Homo sapiens 0-16 22309084-4 2012 Interestingly, the switch from stimulatory to inhibitory actions occurred within a narrow concentration range and correlated with the ability of 15d-PGJ(2) to induce heme oxygenase 1 and gamma-GCSm expression. 15d-pgj 145-152 heme oxygenase 1 Homo sapiens 166-182 22037960-12 2012 Furthermore, we found a significant positive correlation between exhaled CO levels and HO-1 gene expression levels. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 99-103 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Iron 114-118 heme oxygenase 1 Homo sapiens 18-22 22037960-15 2012 These results suggest that HO-1-mediated heme breakdown is caused by I/R during LDLT, since it is associated with increased exhaled CO levels and liver damage. Heme 41-45 heme oxygenase 1 Homo sapiens 27-31 22037960-15 2012 These results suggest that HO-1-mediated heme breakdown is caused by I/R during LDLT, since it is associated with increased exhaled CO levels and liver damage. Carbon Monoxide 144-146 heme oxygenase 1 Homo sapiens 27-31 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 0-16 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 18-22 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Carbon Monoxide 137-139 heme oxygenase 1 Homo sapiens 0-16 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Carbon Monoxide 137-139 heme oxygenase 1 Homo sapiens 18-22 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Biliverdine 158-176 heme oxygenase 1 Homo sapiens 0-16 22037960-3 2012 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme catabolism and results in the production of iron, carbon monoxide (CO), and biliverdin IXalpha. Biliverdine 158-176 heme oxygenase 1 Homo sapiens 18-22 22236404-1 2012 INTRODUCTION: Critical illness leads to increased endogenous production of carbon monoxide (CO) due to the induction of the stress-response enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 148-164 22120038-6 2012 On the other hand, the inhibitory effect of Pb(2+) on the induction of CYP1A1 coincided with an increase in heme oxygenase-1 (HO-1) mRNA level and reactive oxygen species production at the posttranslational level. Lead 44-46 heme oxygenase 1 Homo sapiens 108-124 22120038-6 2012 On the other hand, the inhibitory effect of Pb(2+) on the induction of CYP1A1 coincided with an increase in heme oxygenase-1 (HO-1) mRNA level and reactive oxygen species production at the posttranslational level. Lead 44-46 heme oxygenase 1 Homo sapiens 126-130 21447045-0 2012 Atorvastatin activates heme oxygenase-1 at the stress response elements. Atorvastatin 0-12 heme oxygenase 1 Homo sapiens 23-39 21447045-5 2012 Among the antioxidant response genes, heme oxygenase-1 (HO-1) was significantly up-regulated by atorvastatin. Atorvastatin 96-108 heme oxygenase 1 Homo sapiens 38-54 21447045-5 2012 Among the antioxidant response genes, heme oxygenase-1 (HO-1) was significantly up-regulated by atorvastatin. Atorvastatin 96-108 heme oxygenase 1 Homo sapiens 56-60 21447045-6 2012 Pre-incubation of the cells with geranylgeranyl pyrophosphate blocked atorvastatin-induced apoptosis, but not up-regulation of HO-1, suggesting that atorvastatin-induced apoptosis is dependent on GTPase activity and up-regulation of HO-1 gene is not. geranylgeranyl pyrophosphate 33-61 heme oxygenase 1 Homo sapiens 233-237 21447045-6 2012 Pre-incubation of the cells with geranylgeranyl pyrophosphate blocked atorvastatin-induced apoptosis, but not up-regulation of HO-1, suggesting that atorvastatin-induced apoptosis is dependent on GTPase activity and up-regulation of HO-1 gene is not. Atorvastatin 149-161 heme oxygenase 1 Homo sapiens 233-237 21447045-9 2012 Because these StRE sites are present in clusters in HO-1 promoter, up-regulation of HO-1 by atorvastatin may involve multiple StRE sites. Atorvastatin 92-104 heme oxygenase 1 Homo sapiens 52-56 21447045-9 2012 Because these StRE sites are present in clusters in HO-1 promoter, up-regulation of HO-1 by atorvastatin may involve multiple StRE sites. Atorvastatin 92-104 heme oxygenase 1 Homo sapiens 84-88 21447045-10 2012 The role of HO-1 in atorvastatin-induced apoptosis in PC-3 and MCF-7 remains to be studied. Atorvastatin 20-32 heme oxygenase 1 Homo sapiens 12-16 21447442-0 2012 Epigallocatechin-gallate stimulates NF-E2-related factor and heme oxygenase-1 via caveolin-1 displacement. epigallocatechin gallate 0-24 heme oxygenase 1 Homo sapiens 61-77 21447442-6 2012 To test this hypothesis, we explored the effect of EGCG on the induction of NF-E2-related factor (Nrf2) and HO-1 in endothelial cells with or without functional caveolae. epigallocatechin gallate 51-55 heme oxygenase 1 Homo sapiens 108-112 21447442-7 2012 Treatment with EGCG activated Nrf2 and increased HO-1 expression and cellular production of bilirubin. epigallocatechin gallate 15-19 heme oxygenase 1 Homo sapiens 49-53 22120038-7 2012 Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, or supplementing heme, using hemin, caused a partial restoration of Pb(2+)-mediated inhibition of CYP1A1 induction by TCDD. tin mesoporphyrin 49-66 heme oxygenase 1 Homo sapiens 31-35 22120038-7 2012 Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, or supplementing heme, using hemin, caused a partial restoration of Pb(2+)-mediated inhibition of CYP1A1 induction by TCDD. Hemin 97-102 heme oxygenase 1 Homo sapiens 31-35 22120038-7 2012 Furthermore, the inhibition of HO-1 activity, by tin mesoporphyrin, or supplementing heme, using hemin, caused a partial restoration of Pb(2+)-mediated inhibition of CYP1A1 induction by TCDD. Polychlorinated Dibenzodioxins 186-190 heme oxygenase 1 Homo sapiens 31-35 22120038-8 2012 In addition, transfection of HepG2 cells with siRNA targeting the human HO-1 gene restored the Pb(2+)-mediated inhibition of TCDD-induced CYP1A1. Polychlorinated Dibenzodioxins 125-129 heme oxygenase 1 Homo sapiens 72-76 22158875-9 2012 The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. Reactive Oxygen Species 31-34 heme oxygenase 1 Homo sapiens 68-72 22158875-9 2012 The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. Reactive Oxygen Species 31-34 heme oxygenase 1 Homo sapiens 158-162 22158875-9 2012 The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. Reactive Oxygen Species 139-142 heme oxygenase 1 Homo sapiens 68-72 22158875-9 2012 The IL-19-induced reduction in ROS concentration is attenuated when HO-1 is reduced by siRNA, indicating that the IL-19-driven decrease in ROS is mediated by HO-1 expression. Reactive Oxygen Species 139-142 heme oxygenase 1 Homo sapiens 158-162 22142471-4 2012 In this study, the cancer-chemopreventive agent nordihydroguaiaretic acid (NDGA) increased the level of Nrf2 protein and expression of heme oxygenase-1 (HO-1) in kidney-derived LLC-PK1 and HEK293T cells and in wild-type mouse embryo fibroblasts (MEFs). Masoprocol 48-73 heme oxygenase 1 Homo sapiens 153-157 22142471-4 2012 In this study, the cancer-chemopreventive agent nordihydroguaiaretic acid (NDGA) increased the level of Nrf2 protein and expression of heme oxygenase-1 (HO-1) in kidney-derived LLC-PK1 and HEK293T cells and in wild-type mouse embryo fibroblasts (MEFs). Masoprocol 75-79 heme oxygenase 1 Homo sapiens 153-157 22142471-6 2012 The relevance of the Nrf2/HO-1 axis to antioxidant protection was further demonstrated by the finding that the HO-1 inhibitor stannous-mesoporphyrin abolished protection against hydrogen peroxide conferred by NDGA. stannous mesoporphyrin 126-148 heme oxygenase 1 Homo sapiens 26-30 22142471-6 2012 The relevance of the Nrf2/HO-1 axis to antioxidant protection was further demonstrated by the finding that the HO-1 inhibitor stannous-mesoporphyrin abolished protection against hydrogen peroxide conferred by NDGA. stannous mesoporphyrin 126-148 heme oxygenase 1 Homo sapiens 111-115 22142471-6 2012 The relevance of the Nrf2/HO-1 axis to antioxidant protection was further demonstrated by the finding that the HO-1 inhibitor stannous-mesoporphyrin abolished protection against hydrogen peroxide conferred by NDGA. Hydrogen Peroxide 178-195 heme oxygenase 1 Homo sapiens 26-30 22142471-6 2012 The relevance of the Nrf2/HO-1 axis to antioxidant protection was further demonstrated by the finding that the HO-1 inhibitor stannous-mesoporphyrin abolished protection against hydrogen peroxide conferred by NDGA. Hydrogen Peroxide 178-195 heme oxygenase 1 Homo sapiens 111-115 22142473-0 2012 Resistance of neuroblastoma GI-ME-N cell line to glutathione depletion involves Nrf2 and heme oxygenase-1. Glutathione 49-60 heme oxygenase 1 Homo sapiens 89-105 22142473-3 2012 Here, we show that in GI-ME-N cells, BSO activates Nrf2 and up-regulates heme oxygenase-1 (HO-1). Nitrogen 28-29 heme oxygenase 1 Homo sapiens 73-89 22142473-3 2012 Here, we show that in GI-ME-N cells, BSO activates Nrf2 and up-regulates heme oxygenase-1 (HO-1). Nitrogen 28-29 heme oxygenase 1 Homo sapiens 91-95 22142473-5 2012 Inhibition of HO-1 and silencing of Nrf2 or HO-1 sensitized GI-ME-N cells to BSO, leading to reactive oxygen/nitrogen species overproduction and decreasing viability. reactive oxygen/nitrogen species 93-125 heme oxygenase 1 Homo sapiens 44-48 22142473-6 2012 Moreover, targeting the Nrf2/HO-1 axis sensitized GI-ME-N cells to etoposide more than GSH depletion. gi-me-n 50-57 heme oxygenase 1 Homo sapiens 29-33 22142473-6 2012 Moreover, targeting the Nrf2/HO-1 axis sensitized GI-ME-N cells to etoposide more than GSH depletion. Etoposide 67-76 heme oxygenase 1 Homo sapiens 29-33 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Heme 68-72 heme oxygenase 1 Homo sapiens 48-52 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Heme 68-72 heme oxygenase 1 Homo sapiens 96-100 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Carbon Monoxide 78-80 heme oxygenase 1 Homo sapiens 48-52 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Carbon Monoxide 78-80 heme oxygenase 1 Homo sapiens 96-100 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Bilirubin 85-94 heme oxygenase 1 Homo sapiens 48-52 22138245-10 2012 Taken together with the biochemical function of HO-1 that catalyzes heme into CO and bilirubin, HO-1 expression may improve the circulation and compensate with oxidative tissue damages induced by hypoxia. Bilirubin 85-94 heme oxygenase 1 Homo sapiens 96-100 22052915-4 2012 OBJECTIVE: This study aimed to define the key transcription factor(s) involved in HO-1 induction by heme. Heme 100-104 heme oxygenase 1 Homo sapiens 82-86 22052915-9 2012 Heme-induced HO-1 and LXR-beta were suppressed by knockdown of ATF-1, and HO-1 and LXR-beta were induced by ATF-1 transfection. Heme 0-4 heme oxygenase 1 Homo sapiens 13-17 22052915-9 2012 Heme-induced HO-1 and LXR-beta were suppressed by knockdown of ATF-1, and HO-1 and LXR-beta were induced by ATF-1 transfection. Heme 0-4 heme oxygenase 1 Homo sapiens 74-78 22052915-13 2012 CONCLUSIONS: These results show that ATF-1 mediates HO-1 induction by heme and drives macrophage adaptation to intraplaque hemorrhage. Heme 70-74 heme oxygenase 1 Homo sapiens 52-56 22236404-1 2012 INTRODUCTION: Critical illness leads to increased endogenous production of carbon monoxide (CO) due to the induction of the stress-response enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 75-90 heme oxygenase 1 Homo sapiens 166-170 22236404-1 2012 INTRODUCTION: Critical illness leads to increased endogenous production of carbon monoxide (CO) due to the induction of the stress-response enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 92-94 heme oxygenase 1 Homo sapiens 148-164 22236404-1 2012 INTRODUCTION: Critical illness leads to increased endogenous production of carbon monoxide (CO) due to the induction of the stress-response enzyme, heme oxygenase-1 (HO-1). Carbon Monoxide 92-94 heme oxygenase 1 Homo sapiens 166-170 22195275-0 2012 Heme Oxygenase-1 Attenuates Hypoxia-Induced sFlt-1 and Oxidative Stress in Placental Villi through Its Metabolic Products CO and Bilirubin. Bilirubin 129-138 heme oxygenase 1 Homo sapiens 0-16 22095827-0 2012 Niacin inhibits vascular inflammation via the induction of heme oxygenase-1. Niacin 0-6 heme oxygenase 1 Homo sapiens 59-75 22095827-2 2012 Niacin is a pleiotropic drug that slows the progression of coronary artery disease and increases serum levels of the HO-1 enzymatic product bilirubin. Niacin 0-6 heme oxygenase 1 Homo sapiens 117-121 22095827-2 2012 Niacin is a pleiotropic drug that slows the progression of coronary artery disease and increases serum levels of the HO-1 enzymatic product bilirubin. Bilirubin 140-149 heme oxygenase 1 Homo sapiens 117-121 22095827-3 2012 This study asks if the cardioprotective properties of niacin involve the induction of HO-1. Niacin 54-60 heme oxygenase 1 Homo sapiens 86-90 22095827-8 2012 Treatment of the animals with tin protoporphyrin-IX, a global HO inhibitor, or HO-1 small interfering RNA to knock down carotid artery HO-1 attenuated the ability of niacin to inhibit vascular inflammation. tin protoporphyrin IX 30-51 heme oxygenase 1 Homo sapiens 135-139 22095827-8 2012 Treatment of the animals with tin protoporphyrin-IX, a global HO inhibitor, or HO-1 small interfering RNA to knock down carotid artery HO-1 attenuated the ability of niacin to inhibit vascular inflammation. Niacin 166-172 heme oxygenase 1 Homo sapiens 79-83 22095827-8 2012 Treatment of the animals with tin protoporphyrin-IX, a global HO inhibitor, or HO-1 small interfering RNA to knock down carotid artery HO-1 attenuated the ability of niacin to inhibit vascular inflammation. Niacin 166-172 heme oxygenase 1 Homo sapiens 135-139 22095827-11 2012 CONCLUSIONS: Niacin activates HO-1 in vivo and in vitro. Niacin 13-19 heme oxygenase 1 Homo sapiens 30-34 22701506-5 2012 In addition, 3,4-O-dicaffeoylquinic acid significantly increased the expression of heme oxygenase-1 (HO-1) in HepG2.2.15 cells. caffeoylquinic acid 13-40 heme oxygenase 1 Homo sapiens 83-99 22701506-5 2012 In addition, 3,4-O-dicaffeoylquinic acid significantly increased the expression of heme oxygenase-1 (HO-1) in HepG2.2.15 cells. caffeoylquinic acid 13-40 heme oxygenase 1 Homo sapiens 101-105 22165967-0 2012 The Hsp32 inhibitors SMA-ZnPP and PEG-ZnPP exert major growth-inhibitory effects on D34+/CD38+ and CD34+/CD38- AML progenitor cells. pegylated zinc protoporphyrin 34-42 heme oxygenase 1 Homo sapiens 4-9 22506100-0 2012 Heme oxygenase-1 induction and organic nitrate therapy: beneficial effects on endothelial dysfunction, nitrate tolerance, and vascular oxidative stress. Nitrates 103-110 heme oxygenase 1 Homo sapiens 0-16 22506100-4 2012 We therefore speculated that induction of heme oxygenase-1 (HO-1) could be an efficient strategy to overcome nitrate tolerance and the associated side effects. Nitrates 109-116 heme oxygenase 1 Homo sapiens 42-58 22518279-4 2012 In the present paper, we will discuss the role of heme oxygenase (HO)-1 which is an enzyme of the heme catabolism and cleaves heme to form biliverdin and carbon monoxide (CO). Biliverdine 139-149 heme oxygenase 1 Homo sapiens 50-71 22195275-3 2012 Recently, HO-1 has been shown to downregulate two of these factors, reactive oxygen species and sFlt-1, and we have reported that HO-1 induction attenuates many of the pathological factors of placental ischemia experimentally. Reactive Oxygen Species 68-91 heme oxygenase 1 Homo sapiens 10-14 22518279-4 2012 In the present paper, we will discuss the role of heme oxygenase (HO)-1 which is an enzyme of the heme catabolism and cleaves heme to form biliverdin and carbon monoxide (CO). Carbon Monoxide 154-169 heme oxygenase 1 Homo sapiens 50-71 22506100-4 2012 We therefore speculated that induction of heme oxygenase-1 (HO-1) could be an efficient strategy to overcome nitrate tolerance and the associated side effects. Nitrates 109-116 heme oxygenase 1 Homo sapiens 60-64 22506100-8 2012 With the present paper, we present and discuss our recent and previous findings on the role of HO-1 for the prevention of nitroglycerin-induced nitrate tolerance and for the beneficial effects of PETN therapy. Nitroglycerin 122-135 heme oxygenase 1 Homo sapiens 95-99 22518279-4 2012 In the present paper, we will discuss the role of heme oxygenase (HO)-1 which is an enzyme of the heme catabolism and cleaves heme to form biliverdin and carbon monoxide (CO). Carbon Monoxide 171-173 heme oxygenase 1 Homo sapiens 50-71 22195275-4 2012 Here, we have examined the direct effect of HO-1 and its bioactive metabolites on hypoxia-induced changes in superoxide and sFlt-1 in placental vascular explants and showed that HO-1 and its metabolites attenuate the production of both factors in this system. Superoxides 109-119 heme oxygenase 1 Homo sapiens 44-48 22518279-6 2012 Recent evidence supports the involvement of HO-1 in the antioxidant and antiinflammatory effect of cyclooxygenase(COX)-2-dependent prostacyclin in the vasculature. Epoprostenol 131-143 heme oxygenase 1 Homo sapiens 44-48 22506100-8 2012 With the present paper, we present and discuss our recent and previous findings on the role of HO-1 for the prevention of nitroglycerin-induced nitrate tolerance and for the beneficial effects of PETN therapy. Nitrates 144-151 heme oxygenase 1 Homo sapiens 95-99 22195275-4 2012 Here, we have examined the direct effect of HO-1 and its bioactive metabolites on hypoxia-induced changes in superoxide and sFlt-1 in placental vascular explants and showed that HO-1 and its metabolites attenuate the production of both factors in this system. Superoxides 109-119 heme oxygenase 1 Homo sapiens 178-182 23095372-5 2012 RESULTS: CD163-expressing macrophages, HO-1 and NADPH-p22 expression were located in areas surrounding tubules with iron deposits and filled with erythrocyte casts. Iron 116-120 heme oxygenase 1 Homo sapiens 39-43 22619734-4 2012 Genetic studies have helped identify a number of proteins linking vitamin D to PD pathology, including the major histocompatibility complex (MHC) class II, the vitamin D receptor (VDR), cytochrome P450 2D6 (CYP2D6), chromosome 22, the renin-angiotensin system (RAS), heme oxygenase-1 (HO-1), poly(ADP-ribose) polymerase-1 gene (PARP-1), neurotrophic factor (NTF), and Sp1 transcription factor. Vitamin D 66-75 heme oxygenase 1 Homo sapiens 267-283 22247601-0 2012 Okanin, a chalcone found in the genus Bidens, and 3-penten-2-one inhibit inducible nitric oxide synthase expression via heme oxygenase-1 induction in RAW264.7 macrophages activated with lipopolysaccharide. okanin 0-6 heme oxygenase 1 Homo sapiens 120-136 22247601-0 2012 Okanin, a chalcone found in the genus Bidens, and 3-penten-2-one inhibit inducible nitric oxide synthase expression via heme oxygenase-1 induction in RAW264.7 macrophages activated with lipopolysaccharide. 3-PENTEN-2-ONE 50-64 heme oxygenase 1 Homo sapiens 120-136 22247601-2 2012 Heme oxygenase-1 expression via activation of nuclear factor-erythroid 2-related factor 2 inhibits nitric oxide production and inducible nitric oxide synthase expression in activated macrophages. Nitric Oxide 99-111 heme oxygenase 1 Homo sapiens 0-16 22247601-4 2012 Here, we found that okanin (possessing the alpha-beta unsaturated carbonyl group) induced heme oxygenase-1 expression via nuclear factor-erythroid 2-related factor 2 activation in RAW264.7 macrophages. okanin 20-26 heme oxygenase 1 Homo sapiens 90-106 22247601-5 2012 3-Penten-2-one, of which structure, as in okanin, possesses the alpha-beta unsaturated carbonyl group, also induced nuclear factor-erythroid 2-related factor 2-dependent heme oxygenase-1 expression, while both 2-pentanone (lacking a double bond) and 2-pentene (lacking a carbonyl group) were virtually inactive. 3-PENTEN-2-ONE 0-14 heme oxygenase 1 Homo sapiens 170-186 22247601-5 2012 3-Penten-2-one, of which structure, as in okanin, possesses the alpha-beta unsaturated carbonyl group, also induced nuclear factor-erythroid 2-related factor 2-dependent heme oxygenase-1 expression, while both 2-pentanone (lacking a double bond) and 2-pentene (lacking a carbonyl group) were virtually inactive. okanin 42-48 heme oxygenase 1 Homo sapiens 170-186 22247601-6 2012 In lipopolysaccharide-activated RAW264.7 macrophages, both okanin and 3-penten-2-one inhibited nitric oxide production and inducible nitric oxide synthase expression via heme oxygenase-1 expression. okanin 59-65 heme oxygenase 1 Homo sapiens 170-186 22247601-6 2012 In lipopolysaccharide-activated RAW264.7 macrophages, both okanin and 3-penten-2-one inhibited nitric oxide production and inducible nitric oxide synthase expression via heme oxygenase-1 expression. 3-PENTEN-2-ONE 70-84 heme oxygenase 1 Homo sapiens 170-186 22247601-7 2012 Collectively, our findings suggest that by virtue of its alpha-beta unsaturated carbonyl functional group, okanin can inhibit nitric oxide production and inducible nitric oxide synthase expression via nuclear factor-erythroid 2-related factor 2-dependent heme oxygenase-1 expression in lipopolysaccharide-activated macrophages. okanin 107-113 heme oxygenase 1 Homo sapiens 255-271 22539869-0 2012 Curcumin protects retinal pigment epithelial cells against oxidative stress via induction of heme oxygenase-1 expression and reduction of reactive oxygen. Curcumin 0-8 heme oxygenase 1 Homo sapiens 93-109 22539869-1 2012 PURPOSE: To determine whether curcumin induces expression of the defensive enzyme heme oxygenase-1 (HO-1) and protects cells against oxidative stress in cultured human retinal pigment epithelial cells. Curcumin 30-38 heme oxygenase 1 Homo sapiens 82-98 22539869-1 2012 PURPOSE: To determine whether curcumin induces expression of the defensive enzyme heme oxygenase-1 (HO-1) and protects cells against oxidative stress in cultured human retinal pigment epithelial cells. Curcumin 30-38 heme oxygenase 1 Homo sapiens 100-104 22539869-5 2012 To confirm the protective role of HO-1 in oxidative stress, small interfering RNA (siRNA) against HO-1 or inhibitor of HO-1 was treated with curcumin in retinal pigment epithelium cells. Curcumin 141-149 heme oxygenase 1 Homo sapiens 98-123 22539869-10 2012 Curcumin"s effect on the reduction of ROS was mediated by the increase in HO-1 expression. Curcumin 0-8 heme oxygenase 1 Homo sapiens 74-78 22539869-10 2012 Curcumin"s effect on the reduction of ROS was mediated by the increase in HO-1 expression. Reactive Oxygen Species 38-41 heme oxygenase 1 Homo sapiens 74-78 22539869-11 2012 CONCLUSIONS: Curcumin upregulated the oxidative stress defense enzyme HO-1 and may protect human retinal pigment epithelial cells against oxidative stress by reducing ROS levels. Curcumin 13-21 heme oxygenase 1 Homo sapiens 70-74 23071605-3 2012 In the present study we assessed effect of transgenic expression of human heme oxygenase-1 (hHO-1), an inducible protein capable of cytoprotection by scavenging reactive oxygen species and preventing apoptosis caused by cellular stress during inflammatory processes, in neonatal porcine islet-like cluster cells (NPCCs). Reactive Oxygen Species 161-184 heme oxygenase 1 Homo sapiens 74-90 22246987-10 2012 Expression of Nrf2-dependent anti-oxidant genes including gamma-glutamylcysteine ligase, heme oxygenase-1, and quinone oxidoreductase were down-regulated in MCF-7/ADR cells, and Nrf2 overexpression partially decreased the susceptibility of ALA-hx PDT in MCF-7/ADR cells. Aminolevulinic Acid 240-243 heme oxygenase 1 Homo sapiens 89-105 23209658-0 2012 Desipramine protects neuronal cell death and induces heme oxygenase-1 expression in Mes23.5 dopaminergic neurons. Desipramine 0-11 heme oxygenase 1 Homo sapiens 53-69 23209658-6 2012 Desipramine induces endogenous anti-oxidative enzyme, heme oxygenase-1 (HO-1) protein and mRNA expression in concentration- and time-dependent manners. Desipramine 0-11 heme oxygenase 1 Homo sapiens 54-70 23209658-6 2012 Desipramine induces endogenous anti-oxidative enzyme, heme oxygenase-1 (HO-1) protein and mRNA expression in concentration- and time-dependent manners. Desipramine 0-11 heme oxygenase 1 Homo sapiens 72-76 23209658-7 2012 A different type of antidepressant SSRI (selective serotonin reuptake inhibitor), fluoxetine also shows similar effects of desipramine on HO-1 expression. Fluoxetine 82-92 heme oxygenase 1 Homo sapiens 138-142 23209658-7 2012 A different type of antidepressant SSRI (selective serotonin reuptake inhibitor), fluoxetine also shows similar effects of desipramine on HO-1 expression. Desipramine 123-134 heme oxygenase 1 Homo sapiens 138-142 23209658-8 2012 Moreover, desipramine induces HO-1 expression through activation of ERK and JNK signaling pathways. Desipramine 10-21 heme oxygenase 1 Homo sapiens 30-34 23209658-10 2012 Moreover, desipramine-mediated increase of HO-1 expression is reduced by transfection with siRNA against Nrf2. Desipramine 10-21 heme oxygenase 1 Homo sapiens 43-47 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. zinc protoporphyrin 78-85 heme oxygenase 1 Homo sapiens 27-31 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. zinc protoporphyrin 78-85 heme oxygenase 1 Homo sapiens 46-50 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. Desipramine 128-139 heme oxygenase 1 Homo sapiens 27-31 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. Desipramine 128-139 heme oxygenase 1 Homo sapiens 46-50 23209658-13 2012 Otherwise, activation of HO-1 activity by HO-1 activator and inducer protect 6-OHDA-induced neuronal death. Oxidopamine 77-83 heme oxygenase 1 Homo sapiens 25-29 23209658-13 2012 Otherwise, activation of HO-1 activity by HO-1 activator and inducer protect 6-OHDA-induced neuronal death. Oxidopamine 77-83 heme oxygenase 1 Homo sapiens 42-46 23209658-14 2012 CONCLUSIONS/SIGNIFICANCE: These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Desipramine 54-65 heme oxygenase 1 Homo sapiens 76-80 23071605-3 2012 In the present study we assessed effect of transgenic expression of human heme oxygenase-1 (hHO-1), an inducible protein capable of cytoprotection by scavenging reactive oxygen species and preventing apoptosis caused by cellular stress during inflammatory processes, in neonatal porcine islet-like cluster cells (NPCCs). Reactive Oxygen Species 161-184 heme oxygenase 1 Homo sapiens 92-97 23071605-4 2012 Transduction of NPCCs with adenovirus containing hHO-1 gene significantly reduced apoptosis compared with the GFP-expressing adenovirus control after treatment with either hydrogen peroxide or hTNF-alpha and cycloheximide. Hydrogen Peroxide 172-189 heme oxygenase 1 Homo sapiens 49-54 23071605-4 2012 Transduction of NPCCs with adenovirus containing hHO-1 gene significantly reduced apoptosis compared with the GFP-expressing adenovirus control after treatment with either hydrogen peroxide or hTNF-alpha and cycloheximide. Cycloheximide 208-221 heme oxygenase 1 Homo sapiens 49-54 23071605-5 2012 These protective effects were diminished by co-treatment of hHO-1 antagonist, Zinc protoporphyrin IX. zinc protoporphyrin 78-100 heme oxygenase 1 Homo sapiens 60-65 23071605-9 2012 As expected, fibroblasts derived from the hHO-1 transgenic pigs were significantly resistant to both hydrogen peroxide damage and hTNF-alpha and cycloheximide-mediated apoptosis when compared with wild-type fibroblasts. Hydrogen Peroxide 101-118 heme oxygenase 1 Homo sapiens 42-47 23071605-9 2012 As expected, fibroblasts derived from the hHO-1 transgenic pigs were significantly resistant to both hydrogen peroxide damage and hTNF-alpha and cycloheximide-mediated apoptosis when compared with wild-type fibroblasts. Cycloheximide 145-158 heme oxygenase 1 Homo sapiens 42-47 22514698-5 2012 Inhibition of HO-1 by zinc protoporphyrin IX (ZnPP) sensitized both cell types to the cytotoxicity of chemotherapeutic agents. zinc protoporphyrin 22-44 heme oxygenase 1 Homo sapiens 14-18 22848562-0 2012 Induction of heme oxygenase-1 inhibits cell death in crotonaldehyde-stimulated HepG2 cells via the PKC-delta-p38-Nrf2 pathway. 2-butenal 53-67 heme oxygenase 1 Homo sapiens 13-29 22848562-5 2012 The aim of this study was to examine the effects of HO-1 induction on cell survival in crotonaldehyde-stimulated human hepatocellular carcinoma (HepG2) cells. 2-butenal 87-101 heme oxygenase 1 Homo sapiens 52-56 22848562-6 2012 METHODS: To investigate the signaling pathway involved in crotonaldehyde-induced HO-1 expression, we compared levels of inhibition efficiency of specific inhibitors and specific small interfering RNAs (siRNAs) of several kinases. 2-butenal 58-72 heme oxygenase 1 Homo sapiens 81-85 22848562-9 2012 Treatment with inhibitors of the protein kinase C-delta (PKC-delta) and p38 pathways resulted in obvious blockage of crotonaldehyde-induced HO-1 expression. 2-butenal 117-131 heme oxygenase 1 Homo sapiens 140-144 22848562-10 2012 Furthermore, treatment with HO-1 siRNA and the specific HO-1 inhibitor zinc-protoporphyrin produced an increase in the G(0)/G(1) phase of the cell cycle in crotonaldehyde-stimulated HepG2 cells. protoporphyrin IX 76-90 heme oxygenase 1 Homo sapiens 28-32 22848562-10 2012 Furthermore, treatment with HO-1 siRNA and the specific HO-1 inhibitor zinc-protoporphyrin produced an increase in the G(0)/G(1) phase of the cell cycle in crotonaldehyde-stimulated HepG2 cells. protoporphyrin IX 76-90 heme oxygenase 1 Homo sapiens 56-60 22848562-10 2012 Furthermore, treatment with HO-1 siRNA and the specific HO-1 inhibitor zinc-protoporphyrin produced an increase in the G(0)/G(1) phase of the cell cycle in crotonaldehyde-stimulated HepG2 cells. 2-butenal 156-170 heme oxygenase 1 Homo sapiens 28-32 22848562-10 2012 Furthermore, treatment with HO-1 siRNA and the specific HO-1 inhibitor zinc-protoporphyrin produced an increase in the G(0)/G(1) phase of the cell cycle in crotonaldehyde-stimulated HepG2 cells. 2-butenal 156-170 heme oxygenase 1 Homo sapiens 56-60 22848562-11 2012 CONCLUSIONS: Taken together, the results support an anti-apoptotic role for HO-1 in crotonaldehyde-stimulated human hepatocellular carcinoma cells and provide a mechanism by which induction of HO-1 expression via PKC-delta-p38 MAPK-Nrf2 pathway may promote tumor resistance to oxidative stress. 2-butenal 84-98 heme oxygenase 1 Homo sapiens 76-80 22848562-11 2012 CONCLUSIONS: Taken together, the results support an anti-apoptotic role for HO-1 in crotonaldehyde-stimulated human hepatocellular carcinoma cells and provide a mechanism by which induction of HO-1 expression via PKC-delta-p38 MAPK-Nrf2 pathway may promote tumor resistance to oxidative stress. 2-butenal 84-98 heme oxygenase 1 Homo sapiens 193-197 22514698-5 2012 Inhibition of HO-1 by zinc protoporphyrin IX (ZnPP) sensitized both cell types to the cytotoxicity of chemotherapeutic agents. zinc protoporphyrin 46-50 heme oxygenase 1 Homo sapiens 14-18 22514698-7 2012 Induction of HO-1 protein expression by stannous chloride enhanced the cytoprotection and suppression of apoptosis caused by anticancer agents. stannous chloride 40-57 heme oxygenase 1 Homo sapiens 13-17 22272327-6 2012 In HUVECs exposed to smokers" serum but not to non-smokers" serum we found that oxidative stress increased, whereas nitric oxide and GSH concentrations decreased; interestingly the expression of Nrf2, of heme oxygenase-1 (HO-1) and of glutamate-cysteine ligase catalytic (GCLC) subunit, the rate-limiting step of synthesis of GSH, was decreased. Nitric Oxide 116-128 heme oxygenase 1 Homo sapiens 204-220 22457802-2 2012 The inducible form of the enzyme heme oxygenase, HO-1, which conducts heme degradation, is absent in erythroblasts where hemoglobin (Hb) is synthesized. Heme 33-37 heme oxygenase 1 Homo sapiens 49-53 22457802-3 2012 Yet, the central macrophage, which retains high HO-1 activity, might be suitable to take over degradation of extra, harmful, Hb heme. Heme 128-132 heme oxygenase 1 Homo sapiens 48-52 22879979-0 2012 Heme oxygenase-1 protects retinal endothelial cells against high glucose- and oxidative/nitrosative stress-induced toxicity. Glucose 65-72 heme oxygenase 1 Homo sapiens 0-16 22879979-10 2012 However, a short-time exposure (24 h) to elevated glucose did not alter cell viability, but increased both the levels of intracellular ROS and HO-1 content. Glucose 50-57 heme oxygenase 1 Homo sapiens 143-147 22879979-11 2012 Moreover, the inhibition of HO with SnPPIX unmasked the toxic effect of high glucose and revealed the protection conferred by HO-1. tin protoporphyrin IX 36-42 heme oxygenase 1 Homo sapiens 126-130 22272327-6 2012 In HUVECs exposed to smokers" serum but not to non-smokers" serum we found that oxidative stress increased, whereas nitric oxide and GSH concentrations decreased; interestingly the expression of Nrf2, of heme oxygenase-1 (HO-1) and of glutamate-cysteine ligase catalytic (GCLC) subunit, the rate-limiting step of synthesis of GSH, was decreased. Glutathione 326-329 heme oxygenase 1 Homo sapiens 204-220 22879979-16 2012 Overexpression of HO-1 prevented the toxic effect induced by H(2)O(2) and NOC-18. Water 61-66 heme oxygenase 1 Homo sapiens 18-22 22272327-6 2012 In HUVECs exposed to smokers" serum but not to non-smokers" serum we found that oxidative stress increased, whereas nitric oxide and GSH concentrations decreased; interestingly the expression of Nrf2, of heme oxygenase-1 (HO-1) and of glutamate-cysteine ligase catalytic (GCLC) subunit, the rate-limiting step of synthesis of GSH, was decreased. Glutathione 326-329 heme oxygenase 1 Homo sapiens 222-226 22438807-1 2012 Heme oxygenase 1 (HO-1) is an essential enzyme induced by heme and multiple stimuli associated with critical illness. Heme 58-62 heme oxygenase 1 Homo sapiens 0-16 22438807-1 2012 Heme oxygenase 1 (HO-1) is an essential enzyme induced by heme and multiple stimuli associated with critical illness. Heme 58-62 heme oxygenase 1 Homo sapiens 18-22 22438807-6 2012 Neutrophils were the main HO-1-expressing cells in peripheral blood, and HMOX1 mRNA expression was upregulated by heme-moieties of lysed erythrocytes. Heme 114-118 heme oxygenase 1 Homo sapiens 73-78 22036727-6 2012 The role of HO-1 in cytoprotection against nano-Ag was reinforced by results using pharmacological inducer of HO-1: cobalt protoporphyrin-mediated HO-1 activation in the NRF2i cells prevented nano-Ag-mediated cell death. cobaltiprotoporphyrin 116-137 heme oxygenase 1 Homo sapiens 12-16 22036727-6 2012 The role of HO-1 in cytoprotection against nano-Ag was reinforced by results using pharmacological inducer of HO-1: cobalt protoporphyrin-mediated HO-1 activation in the NRF2i cells prevented nano-Ag-mediated cell death. cobaltiprotoporphyrin 116-137 heme oxygenase 1 Homo sapiens 110-114 22036727-6 2012 The role of HO-1 in cytoprotection against nano-Ag was reinforced by results using pharmacological inducer of HO-1: cobalt protoporphyrin-mediated HO-1 activation in the NRF2i cells prevented nano-Ag-mediated cell death. cobaltiprotoporphyrin 116-137 heme oxygenase 1 Homo sapiens 110-114 21833623-7 2011 Our results indicated that HO-1 counteracts oxidative imbalance reducing ROS levels. ros 73-76 heme oxygenase 1 Homo sapiens 27-31 22001321-11 2011 Inhibitor of HO-1 activity, tin protoporphyrin IX, further increased HA-mediated reactivation of HIV-1 "mini-virus" in Jurkat clones, and this effect was also inhibited by N-acetyl cysteine. tin protoporphyrin IX 28-49 heme oxygenase 1 Homo sapiens 13-17 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. sodium arsenite 31-46 heme oxygenase 1 Homo sapiens 186-202 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. sodium arsenite 31-46 heme oxygenase 1 Homo sapiens 204-208 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. Simvastatin 59-70 heme oxygenase 1 Homo sapiens 186-202 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. Simvastatin 59-70 heme oxygenase 1 Homo sapiens 204-208 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. Lovastatin 75-85 heme oxygenase 1 Homo sapiens 186-202 21910007-1 2011 Treatment of melanoma cells by sodium arsenite or statins (simvastatin and lovastatin) dramatically modified activities of the main cell signaling pathways resulting in the induction of heme oxygenase-1 (HO-1) and in a downregulation of cyclooxygenase-2 (COX-2) protein levels. Lovastatin 75-85 heme oxygenase 1 Homo sapiens 204-208 21910007-4 2011 We demonstrated in the present study that treatment by sodium arsenite or statins with an additional inhibition of HO-1 expression (or activation) caused a substantial upregulation of apoptosis in melanoma cells. sodium arsenite 55-70 heme oxygenase 1 Homo sapiens 115-119 21910007-2 2011 Through heme degradation and the production of carbon monoxide and biliverdin, HO-1 plays a protective role in different scenario of oxidative stress followed by mitochondrial apoptosis. Carbon Monoxide 47-62 heme oxygenase 1 Homo sapiens 79-83 22001321-11 2011 Inhibitor of HO-1 activity, tin protoporphyrin IX, further increased HA-mediated reactivation of HIV-1 "mini-virus" in Jurkat clones, and this effect was also inhibited by N-acetyl cysteine. Acetylcysteine 172-189 heme oxygenase 1 Homo sapiens 13-17 21910007-2 2011 Through heme degradation and the production of carbon monoxide and biliverdin, HO-1 plays a protective role in different scenario of oxidative stress followed by mitochondrial apoptosis. Biliverdine 67-77 heme oxygenase 1 Homo sapiens 79-83 21945498-0 2011 Association of heme oxygenase-1 GT-repeat polymorphism with blood pressure phenotypes and its relevance to future cardiovascular mortality risk: an observation based on arsenic-exposed individuals. Arsenic 169-176 heme oxygenase 1 Homo sapiens 15-31 21840000-2 2011 As statins can stimulate heme oxygenase-1 (HO-1), which increases bilirubin production, we investigated whether statins in routine use increase total bilirubin levels in subjects at high cardiovascular risk. Bilirubin 66-75 heme oxygenase 1 Homo sapiens 25-41 21917385-2 2011 Induction of heme oxygenase-1 (HO-1) in lymphocytes has a similar effect, and it is not likely to be accidental that a key product of HO-1 activity, biliverdin, is homologous to the structure of phycocyanin"s chromophore phycocyanobilin (PhyCB). Biliverdine 149-159 heme oxygenase 1 Homo sapiens 134-138 22466441-0 2011 Ferulic acid induces heme oxygenase-1 via activation of ERK and Nrf2. ferulic acid 0-12 heme oxygenase 1 Homo sapiens 21-37 22466441-1 2011 This study investigated the effect of ferulic acid (FA) on the up-regulation of heme oxygenase-1 (HO-1) in lymphocytes and the molecular mechanisms involved. ferulic acid 38-50 heme oxygenase 1 Homo sapiens 80-96 22466441-1 2011 This study investigated the effect of ferulic acid (FA) on the up-regulation of heme oxygenase-1 (HO-1) in lymphocytes and the molecular mechanisms involved. ferulic acid 38-50 heme oxygenase 1 Homo sapiens 98-102 22466441-6 2011 In addition, lymphocytes treated with FA exhibited activation of extracellular regulated kinase (ERK) and treatments with U0126 (an ERK kinase inhibitor) attenuated the FA induced activation of Nrf2, resulting in a decrease in HO-1 expression. U 0126 122-127 heme oxygenase 1 Homo sapiens 227-231 22466441-7 2011 Zinc protoporphyrin (ZnPP, a HO-1 inhibitor) markedly suppressed cytoprotection from radiation-induced cell damage by FA. zinc protoporphyrin 0-19 heme oxygenase 1 Homo sapiens 29-33 22466441-7 2011 Zinc protoporphyrin (ZnPP, a HO-1 inhibitor) markedly suppressed cytoprotection from radiation-induced cell damage by FA. zinc protoporphyrin 21-25 heme oxygenase 1 Homo sapiens 29-33 21982894-1 2011 Heme oxygenase 1 (HO-1) is a stress-inducible enzyme that degrades redox-active heme-producing biliverdin, carbon monoxide, and Fe(2+). Biliverdine 95-105 heme oxygenase 1 Homo sapiens 0-22 21982894-1 2011 Heme oxygenase 1 (HO-1) is a stress-inducible enzyme that degrades redox-active heme-producing biliverdin, carbon monoxide, and Fe(2+). Carbon Monoxide 107-122 heme oxygenase 1 Homo sapiens 0-22 21982894-1 2011 Heme oxygenase 1 (HO-1) is a stress-inducible enzyme that degrades redox-active heme-producing biliverdin, carbon monoxide, and Fe(2+). ammonium ferrous sulfate 128-134 heme oxygenase 1 Homo sapiens 0-22 21982894-7 2011 Oligonucleotides mimicking miR-155 efficiently inhibited BACH1 protein translation, resulting in a concentration-dependent increase in HMOX1 mRNA and protein expression in human umbilical vein endothelial cells. Oligonucleotides 0-16 heme oxygenase 1 Homo sapiens 135-140 21321939-7 2011 Consistently, overexpression of HO-1 attenuated EV71-induced NADPH oxidase/ROS generation and EV71 replication which were abrogated by pretreatment with an HO-1 inhibitor, zinc protoporphyrin IX (ZnPP IX). Reactive Oxygen Species 75-78 heme oxygenase 1 Homo sapiens 32-36 21321939-7 2011 Consistently, overexpression of HO-1 attenuated EV71-induced NADPH oxidase/ROS generation and EV71 replication which were abrogated by pretreatment with an HO-1 inhibitor, zinc protoporphyrin IX (ZnPP IX). Reactive Oxygen Species 75-78 heme oxygenase 1 Homo sapiens 156-160 21321939-7 2011 Consistently, overexpression of HO-1 attenuated EV71-induced NADPH oxidase/ROS generation and EV71 replication which were abrogated by pretreatment with an HO-1 inhibitor, zinc protoporphyrin IX (ZnPP IX). zinc protoporphyrin 172-194 heme oxygenase 1 Homo sapiens 32-36 21321939-7 2011 Consistently, overexpression of HO-1 attenuated EV71-induced NADPH oxidase/ROS generation and EV71 replication which were abrogated by pretreatment with an HO-1 inhibitor, zinc protoporphyrin IX (ZnPP IX). zinc protoporphyrin 196-203 heme oxygenase 1 Homo sapiens 32-36 21321939-8 2011 Moreover, metabolite of HO-1, carbon monoxide (CO), also diminished ROS formation and EV71 replication which were reversed by pretreatment with a CO scavenger (hemoglobin) and a cyclic GMP-dependent protein kinase (PKG) inhibitor (KT5823). Carbon Monoxide 30-45 heme oxygenase 1 Homo sapiens 24-28 21321939-8 2011 Moreover, metabolite of HO-1, carbon monoxide (CO), also diminished ROS formation and EV71 replication which were reversed by pretreatment with a CO scavenger (hemoglobin) and a cyclic GMP-dependent protein kinase (PKG) inhibitor (KT5823). Carbon Monoxide 47-49 heme oxygenase 1 Homo sapiens 24-28 21321939-8 2011 Moreover, metabolite of HO-1, carbon monoxide (CO), also diminished ROS formation and EV71 replication which were reversed by pretreatment with a CO scavenger (hemoglobin) and a cyclic GMP-dependent protein kinase (PKG) inhibitor (KT5823). Reactive Oxygen Species 68-71 heme oxygenase 1 Homo sapiens 24-28 21917385-4 2011 These considerations suggest that bilirubin, generated within lymphocytes by HO-1 activation, may play a physiological role in the promotion of Treg immunomodulation. Bilirubin 34-43 heme oxygenase 1 Homo sapiens 77-81 21917385-5 2011 This effect of bilirubin is likely to be independent of NADPH oxidase inhibition, since the NAPDH oxidase activity of macrophages is necessary for Treg induction, possibly because it contributes to HO-1 induction in lymphocytes. Bilirubin 15-24 heme oxygenase 1 Homo sapiens 198-202 21963524-0 2011 Manganese potentiates LPS-induced heme-oxygenase 1 in microglia but not dopaminergic cells: role in controlling microglial hydrogen peroxide and inflammatory cytokine output. Manganese 0-9 heme oxygenase 1 Homo sapiens 34-50 21910986-5 2011 Pretreatment with the HO-1 inhibitor, tin protoporphyrin (SnPP), attenuated the inhibitory activities of LA on LPS-induced inflammatory NO, PGE(2), IL-1beta, TNF-alpha, IL-6 and IL-12 production. tin protoporphyrin IX 38-56 heme oxygenase 1 Homo sapiens 22-26 21910986-5 2011 Pretreatment with the HO-1 inhibitor, tin protoporphyrin (SnPP), attenuated the inhibitory activities of LA on LPS-induced inflammatory NO, PGE(2), IL-1beta, TNF-alpha, IL-6 and IL-12 production. S-Nitroso-N-propionyl-D,L-penicillamine 58-62 heme oxygenase 1 Homo sapiens 22-26 21910986-5 2011 Pretreatment with the HO-1 inhibitor, tin protoporphyrin (SnPP), attenuated the inhibitory activities of LA on LPS-induced inflammatory NO, PGE(2), IL-1beta, TNF-alpha, IL-6 and IL-12 production. Prostaglandins E 140-143 heme oxygenase 1 Homo sapiens 22-26 21925249-0 2011 Simvastatin inhibits pro-inflammatory mediators through induction of heme oxygenase-1 expression in lipopolysaccharide-stimulated RAW264.7 macrophages. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 69-85 21925249-3 2011 Here, the possibility that HO-1 is involved in the anti-inflammatory action of simvastatin, using lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages as a model system has been specifically addressed. Simvastatin 79-90 heme oxygenase 1 Homo sapiens 27-31 21925249-4 2011 Our results demonstrated that in the presence of LPS, simvastatin significantly increased HO-1 expression and activity in a dose-dependent manner compared to that of LPS-stimulated alone macrophages. Simvastatin 54-65 heme oxygenase 1 Homo sapiens 90-94 21925249-7 2011 However, these anti-inflammatory activities of simvastatin were markedly reversed by addition of a HO-1 inhibitor zinc protoporphyrin (ZnPP). Simvastatin 47-58 heme oxygenase 1 Homo sapiens 99-103 21925249-7 2011 However, these anti-inflammatory activities of simvastatin were markedly reversed by addition of a HO-1 inhibitor zinc protoporphyrin (ZnPP). zinc protoporphyrin 114-133 heme oxygenase 1 Homo sapiens 99-103 21925249-7 2011 However, these anti-inflammatory activities of simvastatin were markedly reversed by addition of a HO-1 inhibitor zinc protoporphyrin (ZnPP). zinc protoporphyrin 135-139 heme oxygenase 1 Homo sapiens 99-103 21925249-8 2011 Accordingly, the present results indicate that the anti-inflammatory activity of simvastatin could, at least in part, be regulated by induction of HO-1-mediated processes. Simvastatin 81-92 heme oxygenase 1 Homo sapiens 147-151 21910986-0 2011 Anti-inflammatory effects of lindenenyl acetate via heme oxygenase-1 and AMPK in human periodontal ligament cells. lindenenyl acetate 29-47 heme oxygenase 1 Homo sapiens 52-68 21552291-8 2011 Although p53 and its classical targets such as p21 and Mdm2 are activated by both H(2)O(2) and CDDP, we found that the expression of haeme-oxygenase-1 (HO-1)-an antioxidant and antiapoptotic protein-was directly induced only upon H(2)O(2) treatment in a p53-dependent manner. Hydrogen Peroxide 82-90 heme oxygenase 1 Homo sapiens 152-156 21552291-11 2011 Finally, H(2)O(2)-mediated cell death was rescued significantly in p53-deficient cells by antioxidant treatment, as well as by bilirubin, a by-product of HO-1 metabolism. Bilirubin 127-136 heme oxygenase 1 Homo sapiens 154-158 21600248-3 2011 We found SMA-ZnPP showed higher and more efficient (about 2.5 times) intracellular uptake rate than PEG-ZnPP, although both SMA-ZnPP and PEG-ZnPP micelles were localized at endoplasmic reticulum (ER) and inhibited the target enzyme heme oxygenase 1 (HO-1) similarly. peg-znpp 100-108 heme oxygenase 1 Homo sapiens 232-248 21552291-8 2011 Although p53 and its classical targets such as p21 and Mdm2 are activated by both H(2)O(2) and CDDP, we found that the expression of haeme-oxygenase-1 (HO-1)-an antioxidant and antiapoptotic protein-was directly induced only upon H(2)O(2) treatment in a p53-dependent manner. Cisplatin 95-99 heme oxygenase 1 Homo sapiens 152-156 21552291-8 2011 Although p53 and its classical targets such as p21 and Mdm2 are activated by both H(2)O(2) and CDDP, we found that the expression of haeme-oxygenase-1 (HO-1)-an antioxidant and antiapoptotic protein-was directly induced only upon H(2)O(2) treatment in a p53-dependent manner. Hydrogen Peroxide 230-238 heme oxygenase 1 Homo sapiens 133-150 21552291-8 2011 Although p53 and its classical targets such as p21 and Mdm2 are activated by both H(2)O(2) and CDDP, we found that the expression of haeme-oxygenase-1 (HO-1)-an antioxidant and antiapoptotic protein-was directly induced only upon H(2)O(2) treatment in a p53-dependent manner. Hydrogen Peroxide 230-238 heme oxygenase 1 Homo sapiens 152-156 21552291-11 2011 Finally, H(2)O(2)-mediated cell death was rescued significantly in p53-deficient cells by antioxidant treatment, as well as by bilirubin, a by-product of HO-1 metabolism. Hydrogen Peroxide 9-17 heme oxygenase 1 Homo sapiens 154-158 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 0-17 heme oxygenase 1 Homo sapiens 51-67 21868703-0 2011 Heme induces heme oxygenase 1 via Nrf2: role in the homeostatic macrophage response to intraplaque hemorrhage. Heme 0-4 heme oxygenase 1 Homo sapiens 13-29 21868703-7 2011 Challenge of macrophages with purified heme provoked nuclear translocation of Nrf2, and Nrf2 small interfering RNA resulted in significant inhibition of the ability of heme to induce HO-1 protein. Heme 39-43 heme oxygenase 1 Homo sapiens 183-187 21868703-7 2011 Challenge of macrophages with purified heme provoked nuclear translocation of Nrf2, and Nrf2 small interfering RNA resulted in significant inhibition of the ability of heme to induce HO-1 protein. Heme 168-172 heme oxygenase 1 Homo sapiens 183-187 21868703-9 2011 However, an inducible protein transactivator is also probably necessary, as heme-induced HO-1 mRNA expression was fully inhibited by the protein synthesis inhibitor cycloheximide. Cycloheximide 165-178 heme oxygenase 1 Homo sapiens 89-93 22118543-3 2011 Dimethyl sulfoxide (DMSO) completely decreased HO-1 expression in a time-dependent manner, but clearly induced HL-60 cell differentiation, as evidenced by a marked increase in CD11b expression. Dimethyl Sulfoxide 20-24 heme oxygenase 1 Homo sapiens 47-51 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 0-17 heme oxygenase 1 Homo sapiens 69-73 22118543-4 2011 Interestingly, zinc protoporphyrin (ZnPP), a strong inhibitor of HO-1, induced HL-60 cell differentiation. zinc protoporphyrin 15-34 heme oxygenase 1 Homo sapiens 65-69 22118543-4 2011 Interestingly, zinc protoporphyrin (ZnPP), a strong inhibitor of HO-1, induced HL-60 cell differentiation. zinc protoporphyrin 36-40 heme oxygenase 1 Homo sapiens 65-69 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 0-17 heme oxygenase 1 Homo sapiens 106-110 22118543-5 2011 In contrast, treatment with cobalt protoporphyrin (CoPP), an activator of HO-1, decreased CD11b expression. cobaltiprotoporphyrin 28-49 heme oxygenase 1 Homo sapiens 74-78 22118543-5 2011 In contrast, treatment with cobalt protoporphyrin (CoPP), an activator of HO-1, decreased CD11b expression. cobaltiprotoporphyrin 51-55 heme oxygenase 1 Homo sapiens 74-78 22118543-6 2011 Additionally, ZnPP downregulated HO-1 protein expression in HL-60 cells, whereas CoPP induced upregulation. zinc protoporphyrin 14-18 heme oxygenase 1 Homo sapiens 33-37 22118543-7 2011 These results suggest that HO-1 might have a negative function in DMSO-induced HL-60 cell differentiation. Dimethyl Sulfoxide 66-70 heme oxygenase 1 Homo sapiens 27-31 22118543-8 2011 This study provides the first evidence that HO-1 plays an important role in DMSO-induced HL-60 cell differentiation. Dimethyl Sulfoxide 76-80 heme oxygenase 1 Homo sapiens 44-48 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 19-23 heme oxygenase 1 Homo sapiens 51-67 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 19-23 heme oxygenase 1 Homo sapiens 69-73 22118543-0 2011 Dimethylsulfoxide (DMSO) induces downregulation of heme oxygenase-1 (HO-1) in HL-60 cells: involvement of HO-1 in HL-60 cell differentiation. Dimethyl Sulfoxide 19-23 heme oxygenase 1 Homo sapiens 106-110 22118543-3 2011 Dimethyl sulfoxide (DMSO) completely decreased HO-1 expression in a time-dependent manner, but clearly induced HL-60 cell differentiation, as evidenced by a marked increase in CD11b expression. Dimethyl Sulfoxide 0-18 heme oxygenase 1 Homo sapiens 47-51 21840424-4 2011 We first observed that alpha-iso-cubebenol induced HO-1 mRNA and protein expression in a dose- and time-dependent manner via activation of erythroid-specific nuclear factor-regulated factor 2 (Nrf2). alpha-iso-cubebenol 23-42 heme oxygenase 1 Homo sapiens 51-55 21750082-5 2011 RESULTS: In beta-thalassemia the increased production of reactive oxygen species was associated with down-regulation of heme oxygenase-1 and biliverdin reductase and up-regulation of peroxiredoxin-2. Reactive Oxygen Species 57-80 heme oxygenase 1 Homo sapiens 120-136 21840424-7 2011 These results suggested that alpha-iso-cubebenol induced HO-1 expression through the activation of PI3K/Akt, ERK, and Nrf2 signaling. alpha-iso-cubebenol 29-48 heme oxygenase 1 Homo sapiens 57-61 21840424-10 2011 Additionally, treatment with tin-protoporphyrin (SnPP), a selective inhibitor of HO-1, reversed the alpha-iso-cubebenol-mediated inhibition of P. gingivalis LPS-induced pro-inflammatory cytokines. tin protoporphyrin IX 29-47 heme oxygenase 1 Homo sapiens 81-85 21840424-10 2011 Additionally, treatment with tin-protoporphyrin (SnPP), a selective inhibitor of HO-1, reversed the alpha-iso-cubebenol-mediated inhibition of P. gingivalis LPS-induced pro-inflammatory cytokines. S-Nitroso-N-propionyl-D,L-penicillamine 49-53 heme oxygenase 1 Homo sapiens 81-85 21725585-1 2011 Cobalt protoporphyrin (CoPP) is a potent and effective metalloporphyrin inducer of heme oxygenase-1 (HO-1) activity in many tissues. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 83-99 21840424-10 2011 Additionally, treatment with tin-protoporphyrin (SnPP), a selective inhibitor of HO-1, reversed the alpha-iso-cubebenol-mediated inhibition of P. gingivalis LPS-induced pro-inflammatory cytokines. alpha-iso-cubebenol 100-119 heme oxygenase 1 Homo sapiens 81-85 21725585-1 2011 Cobalt protoporphyrin (CoPP) is a potent and effective metalloporphyrin inducer of heme oxygenase-1 (HO-1) activity in many tissues. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 83-99 21840424-11 2011 Hence, alpha-iso-cubebenol might induce anti-inflammatory effects on P. gingivalis LPS-stimulated human THP-1 macrophages by mediating the activation of PI3k/Akt and ERK that leads to over-expression of HO-1 and Nrf-2. alpha-iso-cubebenol 7-26 heme oxygenase 1 Homo sapiens 203-207 21725585-1 2011 Cobalt protoporphyrin (CoPP) is a potent and effective metalloporphyrin inducer of heme oxygenase-1 (HO-1) activity in many tissues. Metalloporphyrins 55-71 heme oxygenase 1 Homo sapiens 83-99 22065904-9 2011 Knockdown of Nrf2 with siRNA attenuated SFN-induced heme oxygenase-1 (HO-1) up-regulation. sulforaphane 40-43 heme oxygenase 1 Homo sapiens 52-68 22168142-2 2011 Heme oxygenase-1 (HO-1) and its derivatives have anti-apoptotic, anti-inflammatory effects and protect against reactive oxygen species, rendering HO-1 a promising molecule to control AVR. Reactive Oxygen Species 111-134 heme oxygenase 1 Homo sapiens 0-16 21735318-6 2011 In addition, we show that combined dopamine and paraquat treatment increases the expression of heme oxygenase-1, an antioxidant response protein. Dopamine 35-43 heme oxygenase 1 Homo sapiens 95-111 21735318-6 2011 In addition, we show that combined dopamine and paraquat treatment increases the expression of heme oxygenase-1, an antioxidant response protein. Paraquat 48-56 heme oxygenase 1 Homo sapiens 95-111 22168142-2 2011 Heme oxygenase-1 (HO-1) and its derivatives have anti-apoptotic, anti-inflammatory effects and protect against reactive oxygen species, rendering HO-1 a promising molecule to control AVR. Reactive Oxygen Species 111-134 heme oxygenase 1 Homo sapiens 18-22 21911884-3 2011 The (GT)n and (TA)n dinucleotide variations in HMOX1 and UGT1A1 gene promoters, respectively, were determined by fragment analysis in all subjects. Dinucleoside Phosphates 20-32 heme oxygenase 1 Homo sapiens 47-52 22168142-11 2011 These effects could be abrogated by the incubation of transgenic PAECs with the specific HO-1 inhibitor zinc protoporphorine IX (Zn(II)PPIX, 20 mum). zinc protoporphorine ix 104-127 heme oxygenase 1 Homo sapiens 89-93 22168142-11 2011 These effects could be abrogated by the incubation of transgenic PAECs with the specific HO-1 inhibitor zinc protoporphorine IX (Zn(II)PPIX, 20 mum). zn(ii)ppix 129-139 heme oxygenase 1 Homo sapiens 89-93 22090784-1 2011 Heme oxygenase-1 (HO-1) system catalyzes heme to biologically active products: carbon monoxide, biliverdin/bilirubin and free iron. Carbon Monoxide 79-94 heme oxygenase 1 Homo sapiens 0-16 22090784-1 2011 Heme oxygenase-1 (HO-1) system catalyzes heme to biologically active products: carbon monoxide, biliverdin/bilirubin and free iron. Biliverdine 96-106 heme oxygenase 1 Homo sapiens 0-16 22090784-1 2011 Heme oxygenase-1 (HO-1) system catalyzes heme to biologically active products: carbon monoxide, biliverdin/bilirubin and free iron. Bilirubin 107-116 heme oxygenase 1 Homo sapiens 0-16 22090784-1 2011 Heme oxygenase-1 (HO-1) system catalyzes heme to biologically active products: carbon monoxide, biliverdin/bilirubin and free iron. Iron 126-130 heme oxygenase 1 Homo sapiens 0-16 21380847-6 2011 Furthermore, our results show that the hormetic effects of low levels of curcumin are achieved by virtue of it being a hormetin in terms of the induction of stress response pathways, including Nrf2 and HO-1 in human cells. Curcumin 73-81 heme oxygenase 1 Homo sapiens 202-206 21091076-3 2011 By degrading the oxidant heme and generating the antioxidant bilirubin and anti-inflammatory molecule carbon monoxide, HO-1 may protect cell from injury due to oxidative and pathological stress. Heme 25-29 heme oxygenase 1 Homo sapiens 119-123 21091076-3 2011 By degrading the oxidant heme and generating the antioxidant bilirubin and anti-inflammatory molecule carbon monoxide, HO-1 may protect cell from injury due to oxidative and pathological stress. Bilirubin 61-70 heme oxygenase 1 Homo sapiens 119-123 21091076-3 2011 By degrading the oxidant heme and generating the antioxidant bilirubin and anti-inflammatory molecule carbon monoxide, HO-1 may protect cell from injury due to oxidative and pathological stress. Carbon Monoxide 102-117 heme oxygenase 1 Homo sapiens 119-123 21950555-8 2011 Inhibitor of HO-1, Znpp, can increase the radiosensitivity of human NSCLC A549 cells. zinc protoporphyrin 19-23 heme oxygenase 1 Homo sapiens 13-17 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 0-28 heme oxygenase 1 Homo sapiens 110-126 21950555-2 2011 The aim of this study was to evaluate whether Inhibitor of HO-1, zinc protoporphyrin IX (Znpp), enhances the radiosensitivity in human nonsmall cell lung cancer (NSCLC) A549 Cells. zinc protoporphyrin 89-93 heme oxygenase 1 Homo sapiens 59-87 21950555-6 2011 First, overexpression of the HO-1 mRNA was found in treatment with irradiation alone in A549 cells, and expression of the HO-1 mRNA was reduced after combined treatments with 12 mumol/L of Znpp and irradiation. zinc protoporphyrin 189-193 heme oxygenase 1 Homo sapiens 29-33 21950555-6 2011 First, overexpression of the HO-1 mRNA was found in treatment with irradiation alone in A549 cells, and expression of the HO-1 mRNA was reduced after combined treatments with 12 mumol/L of Znpp and irradiation. zinc protoporphyrin 189-193 heme oxygenase 1 Homo sapiens 122-126 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Heme 68-72 heme oxygenase 1 Homo sapiens 12-28 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Heme 68-72 heme oxygenase 1 Homo sapiens 30-34 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Carbon Monoxide 78-93 heme oxygenase 1 Homo sapiens 12-28 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Carbon Monoxide 78-93 heme oxygenase 1 Homo sapiens 30-34 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 12-28 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 30-34 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 12-28 22088544-1 2011 BACKGROUND: Heme oxygenase-1 (HO-1) is an enzyme, which catabolizes heme into carbon monoxide, biliverdin and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 30-34 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 0-28 heme oxygenase 1 Homo sapiens 128-132 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 30-34 heme oxygenase 1 Homo sapiens 110-126 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 30-34 heme oxygenase 1 Homo sapiens 128-132 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Nitrates 21-28 heme oxygenase 1 Homo sapiens 110-126 21844097-2 2011 Pentaerithrityl tetranitrate (PETN) is an organic nitrate with potent antioxidant properties via induction of heme oxygenase-1 (HO-1). Nitrates 21-28 heme oxygenase 1 Homo sapiens 128-132 21844097-13 2011 The expression of the antioxidant enzyme, HO-1, was increased by STZ treatment and further upregulated by PETN, but not ISMN, via activation of the transcription factor NRF2. Streptozocin 65-68 heme oxygenase 1 Homo sapiens 42-46 21902835-2 2011 Heme oxygenase-1 (HMOX1) is an essential enzyme in heme catabolism that is induced by oxidative stress and may play a protective role as an antioxidant in the lung. Heme 51-55 heme oxygenase 1 Homo sapiens 0-16 21499987-3 2011 Data from our and other laboratories have previously demonstrated that curcumin, the yellow pigment of curry, strongly induces heme-oxygenase-1 (HO-1) expression and activity in different brain cells via the activation of heterodimers of NF-E2-related factors 2 (Nrf2)/antioxidant responsive element (ARE) pathway. Curcumin 71-79 heme oxygenase 1 Homo sapiens 127-143 21499987-3 2011 Data from our and other laboratories have previously demonstrated that curcumin, the yellow pigment of curry, strongly induces heme-oxygenase-1 (HO-1) expression and activity in different brain cells via the activation of heterodimers of NF-E2-related factors 2 (Nrf2)/antioxidant responsive element (ARE) pathway. Curcumin 71-79 heme oxygenase 1 Homo sapiens 145-149 21499987-6 2011 Recent and unpublished data from our group revealed that low concentrations of epigallocatechin-3-gallate, the major green tea catechin, induces HO-1 by ARE/Nrf2 pathway in hippocampal neurons, and by this induction, it is able to protect neurons against different models of oxidative damages. epigallocatechin gallate 79-105 heme oxygenase 1 Homo sapiens 145-149 21499987-6 2011 Recent and unpublished data from our group revealed that low concentrations of epigallocatechin-3-gallate, the major green tea catechin, induces HO-1 by ARE/Nrf2 pathway in hippocampal neurons, and by this induction, it is able to protect neurons against different models of oxidative damages. Catechin 87-95 heme oxygenase 1 Homo sapiens 145-149 21499987-7 2011 Furthermore, we have demonstrated that other phenolics, such as caffeic acid phenethyl ester and ethyl ferulate, are also able to protect neurons via HO-1 induction. caffeic acid phenethyl ester 64-92 heme oxygenase 1 Homo sapiens 150-154 21499987-7 2011 Furthermore, we have demonstrated that other phenolics, such as caffeic acid phenethyl ester and ethyl ferulate, are also able to protect neurons via HO-1 induction. ethyl ferulate 97-111 heme oxygenase 1 Homo sapiens 150-154 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 79-84 heme oxygenase 1 Homo sapiens 0-16 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 79-84 heme oxygenase 1 Homo sapiens 18-22 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 201-206 heme oxygenase 1 Homo sapiens 0-16 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 201-206 heme oxygenase 1 Homo sapiens 18-22 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 201-206 heme oxygenase 1 Homo sapiens 0-16 21681819-3 2011 Heme oxygenase-1 (HO-1) expression was first evident after 2 h incubation with hemin, with maximal expression being observed by 24 h. Despite the induction of HO-1, it was found that the proportion of hemin metabolized by astrocytes remained fairly constant throughout the 24 h period, with 70-80% of intracellular hemin remaining intact. Hemin 201-206 heme oxygenase 1 Homo sapiens 18-22 22008538-9 2011 Auranofin-inducted HO-1 gene expression was observed in human primary hepatocytes as well as mouse primary hepatocytes. Auranofin 0-9 heme oxygenase 1 Homo sapiens 19-23 21902835-2 2011 Heme oxygenase-1 (HMOX1) is an essential enzyme in heme catabolism that is induced by oxidative stress and may play a protective role as an antioxidant in the lung. Heme 51-55 heme oxygenase 1 Homo sapiens 18-23 21741353-1 2011 Genetic variations in POR, encoding NADPH-cytochrome P450 oxidoreductase (CYPOR), can diminish the function of numerous cytochromes P450, and also have the potential to block degradation of heme by heme oxygenase-1 (HO-1). Heme 190-194 heme oxygenase 1 Homo sapiens 198-214 21741353-1 2011 Genetic variations in POR, encoding NADPH-cytochrome P450 oxidoreductase (CYPOR), can diminish the function of numerous cytochromes P450, and also have the potential to block degradation of heme by heme oxygenase-1 (HO-1). Heme 190-194 heme oxygenase 1 Homo sapiens 216-220 21741353-6 2011 When mixed with WT CYPOR, only the Y181D CYPOR variant inhibited heme degradation by sequestering HO-1, whereas Y459H and V492E were unable to inhibit HO-1 activity suggesting that CYPOR variants might have differential binding affinities with redox partners. Heme 65-69 heme oxygenase 1 Homo sapiens 98-102 21669191-5 2011 It was found that the electron-withdrawing (NO(2), CN, halogen) groups in BMN molecules and double meta-MeO substituents increased the HO-1 gene induction, while the electron-donating groups in ortho/para position (OH, MeO and N-morpholino) significantly decreased it. Halogens 55-62 heme oxygenase 1 Homo sapiens 135-139 21975817-1 2011 In the present study, we investigated an anti-inflammatory effect of ethyl gallate (EG) isolated from Galla Rhois as evaluated by inhibition of nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression, and a potential role of heme oxygenase-1 (HO-1) in the inhibition of NO production elicited by EG. ethyl gallate 69-82 heme oxygenase 1 Homo sapiens 252-268 21975817-1 2011 In the present study, we investigated an anti-inflammatory effect of ethyl gallate (EG) isolated from Galla Rhois as evaluated by inhibition of nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression, and a potential role of heme oxygenase-1 (HO-1) in the inhibition of NO production elicited by EG. ethyl gallate 69-82 heme oxygenase 1 Homo sapiens 270-274 21975817-1 2011 In the present study, we investigated an anti-inflammatory effect of ethyl gallate (EG) isolated from Galla Rhois as evaluated by inhibition of nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression, and a potential role of heme oxygenase-1 (HO-1) in the inhibition of NO production elicited by EG. ethyl gallate 84-86 heme oxygenase 1 Homo sapiens 252-268 21975817-1 2011 In the present study, we investigated an anti-inflammatory effect of ethyl gallate (EG) isolated from Galla Rhois as evaluated by inhibition of nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) expression, and a potential role of heme oxygenase-1 (HO-1) in the inhibition of NO production elicited by EG. ethyl gallate 84-86 heme oxygenase 1 Homo sapiens 270-274 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. S-Nitroso-N-propionyl-D,L-penicillamine 28-32 heme oxygenase 1 Homo sapiens 70-74 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. S-Nitroso-N-propionyl-D,L-penicillamine 28-32 heme oxygenase 1 Homo sapiens 166-170 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. tin protoporphyrin IX 34-55 heme oxygenase 1 Homo sapiens 70-74 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. tin protoporphyrin IX 34-55 heme oxygenase 1 Homo sapiens 166-170 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. ethyl gallate 124-126 heme oxygenase 1 Homo sapiens 70-74 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. ethyl gallate 124-126 heme oxygenase 1 Homo sapiens 166-170 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. Nitrites 130-137 heme oxygenase 1 Homo sapiens 166-170 21975817-5 2011 In addition, treatment with SnPP (tin protoporphyrin IX), a selective HO-1 inhibitor, counteracted the inhibitory effect of EG on nitrite production, suggesting that HO-1 is, at least in part, implicated in the inhibition of NO production induced by EG treatment. ethyl gallate 250-252 heme oxygenase 1 Homo sapiens 166-170 21975817-0 2011 Involvement of heme oxygenase-1 induction in inhibitory effect of ethyl gallate isolated from Galla Rhois on nitric oxide production in RAW 264.7 macrophages. ethyl gallate 66-79 heme oxygenase 1 Homo sapiens 15-31 21975817-0 2011 Involvement of heme oxygenase-1 induction in inhibitory effect of ethyl gallate isolated from Galla Rhois on nitric oxide production in RAW 264.7 macrophages. Nitric Oxide 109-121 heme oxygenase 1 Homo sapiens 15-31 21669191-5 2011 It was found that the electron-withdrawing (NO(2), CN, halogen) groups in BMN molecules and double meta-MeO substituents increased the HO-1 gene induction, while the electron-donating groups in ortho/para position (OH, MeO and N-morpholino) significantly decreased it. bmn 74-77 heme oxygenase 1 Homo sapiens 135-139 21669191-5 2011 It was found that the electron-withdrawing (NO(2), CN, halogen) groups in BMN molecules and double meta-MeO substituents increased the HO-1 gene induction, while the electron-donating groups in ortho/para position (OH, MeO and N-morpholino) significantly decreased it. meta- 99-104 heme oxygenase 1 Homo sapiens 135-139 21669191-5 2011 It was found that the electron-withdrawing (NO(2), CN, halogen) groups in BMN molecules and double meta-MeO substituents increased the HO-1 gene induction, while the electron-donating groups in ortho/para position (OH, MeO and N-morpholino) significantly decreased it. n-morpholino 227-239 heme oxygenase 1 Homo sapiens 135-139 21620964-0 2011 An IkappaBalpha phosphorylation inhibitor induces heme oxygenase-1(HO-1) expression through the activation of reactive oxygen species (ROS)-Nrf2-ARE signaling and ROS-PI3K/Akt signaling in an NF-kappaB-independent mechanism. Reactive Oxygen Species 110-133 heme oxygenase 1 Homo sapiens 50-71 21843792-4 2011 Additionally, PGEA inhibited the TNF-alpha/IFN-gamma-co-induced activation of NF-kappaB and STAT1 and increased the expression of heme oxygenase-1 (HO-1) protein and mRNA. pgea 14-18 heme oxygenase 1 Homo sapiens 130-146 21843792-4 2011 Additionally, PGEA inhibited the TNF-alpha/IFN-gamma-co-induced activation of NF-kappaB and STAT1 and increased the expression of heme oxygenase-1 (HO-1) protein and mRNA. pgea 14-18 heme oxygenase 1 Homo sapiens 148-152 21843792-5 2011 HO-1 inhibitor enhanced the suppressive effects of PGEA on TNF-alpha/IFN-gamma-co-induced TARC production and gene expression. pgea 51-55 heme oxygenase 1 Homo sapiens 0-4 21843792-6 2011 Collectively, these data demonstrate that PGEA inhibits chemokine expression in keratinocytes by inducing HO-1 expression and it suggests a possible therapeutic application in atopic dermatitis and other inflammatory skin diseases. pgea 42-46 heme oxygenase 1 Homo sapiens 106-110 21520244-0 2011 Fisetin induces Nrf2-mediated HO-1 expression through PKC-delta and p38 in human umbilical vein endothelial cells. fisetin 0-7 heme oxygenase 1 Homo sapiens 30-34 21520244-3 2011 In the present study, we investigated the effect of fisetin on the up-regulation of HO-1 in human umbilical vein endothelial cells (HUVECs). fisetin 52-59 heme oxygenase 1 Homo sapiens 84-88 21520244-5 2011 Fisetin treatment resulted in significantly increased NF-E2-related factor 2 (Nrf2) nuclear translocation, and antioxidant response element (ARE)-luciferase activity, leading to up-regulation of HO-1 expression. fisetin 0-7 heme oxygenase 1 Homo sapiens 195-199 21520244-6 2011 In addition, fisetin pretreatment reduced hydrogen peroxide (H(2)O(2))-induced cell death, and this effect was reversed by ZnPP, an inhibitor of HO-1. fisetin 13-20 heme oxygenase 1 Homo sapiens 145-149 21520244-6 2011 In addition, fisetin pretreatment reduced hydrogen peroxide (H(2)O(2))-induced cell death, and this effect was reversed by ZnPP, an inhibitor of HO-1. Hydrogen Peroxide 42-59 heme oxygenase 1 Homo sapiens 145-149 21520244-6 2011 In addition, fisetin pretreatment reduced hydrogen peroxide (H(2)O(2))-induced cell death, and this effect was reversed by ZnPP, an inhibitor of HO-1. Hydrogen Peroxide 61-69 heme oxygenase 1 Homo sapiens 145-149 21520244-6 2011 In addition, fisetin pretreatment reduced hydrogen peroxide (H(2)O(2))-induced cell death, and this effect was reversed by ZnPP, an inhibitor of HO-1. zinc protoporphyrin 123-127 heme oxygenase 1 Homo sapiens 145-149 21520244-7 2011 In summary, these findings suggest that induction of HO-1 expression via Nrf2 activation may contribute to the cytoprotection exerted by fisetin against H(2)O(2) -induced oxidative stress in HUVECs. Hydrogen Peroxide 153-161 heme oxygenase 1 Homo sapiens 53-57 21620964-0 2011 An IkappaBalpha phosphorylation inhibitor induces heme oxygenase-1(HO-1) expression through the activation of reactive oxygen species (ROS)-Nrf2-ARE signaling and ROS-PI3K/Akt signaling in an NF-kappaB-independent mechanism. Reactive Oxygen Species 135-138 heme oxygenase 1 Homo sapiens 50-71 21620964-0 2011 An IkappaBalpha phosphorylation inhibitor induces heme oxygenase-1(HO-1) expression through the activation of reactive oxygen species (ROS)-Nrf2-ARE signaling and ROS-PI3K/Akt signaling in an NF-kappaB-independent mechanism. Reactive Oxygen Species 163-166 heme oxygenase 1 Homo sapiens 50-71 21620964-3 2011 Antioxidant enzymes, such as, heme oxygenase-1 (HO-1), tightly regulate ROS levels within cells. Reactive Oxygen Species 72-75 heme oxygenase 1 Homo sapiens 30-46 21620964-3 2011 Antioxidant enzymes, such as, heme oxygenase-1 (HO-1), tightly regulate ROS levels within cells. Reactive Oxygen Species 72-75 heme oxygenase 1 Homo sapiens 48-52 21620964-6 2011 In addition, PI3K/Akt inhibitor (LY294002) blocked Bay-induced HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 33-41 heme oxygenase 1 Homo sapiens 63-67 21620964-7 2011 Pretreatment with anti-oxidants (N-acetylcysteine (NAC) or glutathione) significantly reduced Bay-induced HO-1 mRNA/protein expression, nuclear translocation of Nrf2 and phosphorylation of Akt. Acetylcysteine 33-49 heme oxygenase 1 Homo sapiens 106-110 21620964-7 2011 Pretreatment with anti-oxidants (N-acetylcysteine (NAC) or glutathione) significantly reduced Bay-induced HO-1 mRNA/protein expression, nuclear translocation of Nrf2 and phosphorylation of Akt. Acetylcysteine 51-54 heme oxygenase 1 Homo sapiens 106-110 21620964-7 2011 Pretreatment with anti-oxidants (N-acetylcysteine (NAC) or glutathione) significantly reduced Bay-induced HO-1 mRNA/protein expression, nuclear translocation of Nrf2 and phosphorylation of Akt. Glutathione 59-70 heme oxygenase 1 Homo sapiens 106-110 21620964-9 2011 Furthermore, other NF-kappaB inhibitors, such as pyrrolidine dithiocarbamate (PDTC) and MG132, also increased HO-1 mRNA and protein expression. pyrrolidine dithiocarbamic acid 49-76 heme oxygenase 1 Homo sapiens 110-114 21620964-9 2011 Furthermore, other NF-kappaB inhibitors, such as pyrrolidine dithiocarbamate (PDTC) and MG132, also increased HO-1 mRNA and protein expression. pyrrolidine dithiocarbamic acid 78-82 heme oxygenase 1 Homo sapiens 110-114 21620964-9 2011 Furthermore, other NF-kappaB inhibitors, such as pyrrolidine dithiocarbamate (PDTC) and MG132, also increased HO-1 mRNA and protein expression. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 88-93 heme oxygenase 1 Homo sapiens 110-114 21620964-11 2011 Taken together, our results suggest that in human colon cancer HT29 cells, Bay induces HO-1 expression by increasing ROS production in an Nrf2-ARE and PI3K dependent manner, but Bay acts independently of NF-kappaB. Reactive Oxygen Species 117-120 heme oxygenase 1 Homo sapiens 87-91 21327864-0 2011 Inhibition of heme oxygenase-1 enhances anti-cancer effects of arsenic trioxide on glioma cells. Arsenic Trioxide 63-79 heme oxygenase 1 Homo sapiens 14-30 21327864-2 2011 In this study we showed that ATO induced cell damage and heme oxygenase-1 (HO-1) expression in glioma cells via ROS generation. Arsenic Trioxide 29-32 heme oxygenase 1 Homo sapiens 57-73 21327864-2 2011 In this study we showed that ATO induced cell damage and heme oxygenase-1 (HO-1) expression in glioma cells via ROS generation. Arsenic Trioxide 29-32 heme oxygenase 1 Homo sapiens 75-79 21327864-2 2011 In this study we showed that ATO induced cell damage and heme oxygenase-1 (HO-1) expression in glioma cells via ROS generation. ros 112-115 heme oxygenase 1 Homo sapiens 75-79 21327864-3 2011 HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. Arsenic Trioxide 36-39 heme oxygenase 1 Homo sapiens 0-4 21327864-3 2011 HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. ros 63-66 heme oxygenase 1 Homo sapiens 0-4 21327864-3 2011 HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. ros 146-149 heme oxygenase 1 Homo sapiens 83-87 21327864-3 2011 HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. Arsenic Trioxide 172-175 heme oxygenase 1 Homo sapiens 0-4 21327864-3 2011 HO-1 inducer clearly protected from ATO-induced cell death and ROS generation, and HO-1 inhibitor led to a significant increase in cell death and ROS generation induced by ATO. Arsenic Trioxide 172-175 heme oxygenase 1 Homo sapiens 83-87 21327864-4 2011 In addition, knockdown of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) strongly inhibited HO-1 expression induced by ATO, and significantly enhanced ATO-induced oxidative damage. Arsenic Trioxide 123-126 heme oxygenase 1 Homo sapiens 96-100 21327864-6 2011 Considering that HO-1 is highly expressed in glioma tissues, administration of ATO in combination with either HO-1 inhibitor or Nrf2 knockdown may act as a new approach to the treatment of glioma. Arsenic Trioxide 79-82 heme oxygenase 1 Homo sapiens 17-21 21533649-0 2011 Dimethyl sulfoxide induces heme oxygenase-1 expression via JNKs and Nrf2 pathways in human umbilical vein endothelial cells. Dimethyl Sulfoxide 0-18 heme oxygenase 1 Homo sapiens 27-43 21533649-3 2011 In this paper, we investigated the capability of DMSO to up-regulate heme oxygenase-1(HO-1) expression, as well as the possible underlying mechanisms in human umbilical vein endothelial cells (HUVECs). Dimethyl Sulfoxide 49-53 heme oxygenase 1 Homo sapiens 69-85 21911919-4 2011 This study aims to analyse the role of HO-1 in regulating apoptosis in AML cells in response to two front-line chemotherapeutic agents used for AML, cytarabine and daunorubicin. Cytarabine 149-159 heme oxygenase 1 Homo sapiens 39-43 21640381-0 2011 Carbon monoxide, a reaction product of heme oxygenase-1, suppresses the expression of C-reactive protein by endoplasmic reticulum stress through modulation of the unfolded protein response. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 39-55 21668870-6 2011 The ALA-dependent accumulation of protoporphyrin was decreased in HeLa cells by transfection with HO-1 and HO-2 cDNA. Aminolevulinic Acid 4-7 heme oxygenase 1 Homo sapiens 98-111 21668870-6 2011 The ALA-dependent accumulation of protoporphyrin was decreased in HeLa cells by transfection with HO-1 and HO-2 cDNA. protoporphyrin IX 34-48 heme oxygenase 1 Homo sapiens 98-111 21911919-4 2011 This study aims to analyse the role of HO-1 in regulating apoptosis in AML cells in response to two front-line chemotherapeutic agents used for AML, cytarabine and daunorubicin. Daunorubicin 164-176 heme oxygenase 1 Homo sapiens 39-43 21911919-5 2011 Here we show that HO-1 expression in AML samples was increased in response to both cytarabine and daunorubicin treatment, and micro RNA (miRNA) silenced HO-1 expression in combination with either daunorubicin or cytarabine induced a greater apoptotic responses in AML cells. Cytarabine 83-93 heme oxygenase 1 Homo sapiens 18-22 21911919-5 2011 Here we show that HO-1 expression in AML samples was increased in response to both cytarabine and daunorubicin treatment, and micro RNA (miRNA) silenced HO-1 expression in combination with either daunorubicin or cytarabine induced a greater apoptotic responses in AML cells. Daunorubicin 98-110 heme oxygenase 1 Homo sapiens 18-22 21911919-5 2011 Here we show that HO-1 expression in AML samples was increased in response to both cytarabine and daunorubicin treatment, and micro RNA (miRNA) silenced HO-1 expression in combination with either daunorubicin or cytarabine induced a greater apoptotic responses in AML cells. Cytarabine 212-222 heme oxygenase 1 Homo sapiens 153-157 21911919-7 2011 However, ROS-dependent induction of HO-1 was limiting the apoptotic response that is seen in AML towards cytarabine and daunorubicin treatment. Reactive Oxygen Species 9-12 heme oxygenase 1 Homo sapiens 36-40 21911919-7 2011 However, ROS-dependent induction of HO-1 was limiting the apoptotic response that is seen in AML towards cytarabine and daunorubicin treatment. Cytarabine 105-115 heme oxygenase 1 Homo sapiens 36-40 21911919-7 2011 However, ROS-dependent induction of HO-1 was limiting the apoptotic response that is seen in AML towards cytarabine and daunorubicin treatment. Daunorubicin 120-132 heme oxygenase 1 Homo sapiens 36-40 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Glutathione 19-30 heme oxygenase 1 Homo sapiens 122-126 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Glutathione 19-30 heme oxygenase 1 Homo sapiens 220-224 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Acetylcysteine 37-56 heme oxygenase 1 Homo sapiens 122-126 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Acetylcysteine 37-56 heme oxygenase 1 Homo sapiens 220-224 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Carbon Monoxide 274-276 heme oxygenase 1 Homo sapiens 122-126 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Biliverdine 278-288 heme oxygenase 1 Homo sapiens 122-126 21635873-5 2011 Interestingly, the glutathione donor N-acetyl-l-cysteine or the NADPH oxidase inhibitor apocynin blocked the induction of HO-1 by compound C. Finally, compound C stimulated EC death and this was potentiated by silencing HO-1 expression and reversed by the administration of CO, biliverdin, or bilirubin. Bilirubin 293-302 heme oxygenase 1 Homo sapiens 122-126 20094823-12 2011 The analysis reveals that eCO levels were significantly raised in asthma and it may attribute to high expression of HO-1, but there were no significantly high eCO levels between AR and control groups. ECO 26-29 heme oxygenase 1 Homo sapiens 116-120 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Heme 17-21 heme oxygenase 1 Homo sapiens 35-38 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Carbon Monoxide 108-110 heme oxygenase 1 Homo sapiens 17-33 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Carbon Monoxide 108-110 heme oxygenase 1 Homo sapiens 35-38 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Biliverdine 96-106 heme oxygenase 1 Homo sapiens 17-33 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Iron 123-127 heme oxygenase 1 Homo sapiens 17-33 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Biliverdine 96-106 heme oxygenase 1 Homo sapiens 35-38 21079975-3 2011 Intracellularly, heme oxygenase-1 (HO1) participates in the cleavage of the heme ring producing biliverdin, CO and ferrous iron. Iron 123-127 heme oxygenase 1 Homo sapiens 35-38 22053693-6 2011 The ROS promotes lipid oxidation and are known to induce stress proteins, such as hemeoxygenase 1 (HO-1) and heat-shock protein 70 (HSP70). Reactive Oxygen Species 4-7 heme oxygenase 1 Homo sapiens 82-97 21079975-6 2011 OBJECTIVE: This study focused on the uptake and transport of heme iron and on the role of heme oxygenase-1 on heme iron metabolism. Iron 115-119 heme oxygenase 1 Homo sapiens 90-106 21079975-8 2011 A full-length heme oxygenase-1 cDNA was expressed in Caco-2 cells and intracellular iron and heme-Fe content, heme uptake, heme and iron transport and heme oxygenase-1 immunolocalization were assessed in these cells. Iron 84-88 heme oxygenase 1 Homo sapiens 14-30 21079975-9 2011 RESULTS: Heme-Fe was bioavailable and induced an intracellular increase in iron, ferritin and HO1 levels and a decrease in DMT1 expression. heme-fe 9-16 heme oxygenase 1 Homo sapiens 94-97 21079975-10 2011 In cells overexpressing HO1, heme-Fe uptake and transepithelial Fe transport was higher than in controls. Heme 29-33 heme oxygenase 1 Homo sapiens 24-27 21079975-10 2011 In cells overexpressing HO1, heme-Fe uptake and transepithelial Fe transport was higher than in controls. Iron 34-36 heme oxygenase 1 Homo sapiens 24-27 21079975-10 2011 In cells overexpressing HO1, heme-Fe uptake and transepithelial Fe transport was higher than in controls. Iron 64-66 heme oxygenase 1 Homo sapiens 24-27 21079975-13 2011 In a high heme-Fe condition, HO1 is found near the plasma membrane. Heme 10-14 heme oxygenase 1 Homo sapiens 29-32 21079975-14 2011 CONCLUSIONS: These results suggest that heme oxygenase-1 catabolizes most of the heme-Fe and favors iron influx and efflux in intestinal cells. heme-fe 81-88 heme oxygenase 1 Homo sapiens 40-56 21079975-14 2011 CONCLUSIONS: These results suggest that heme oxygenase-1 catabolizes most of the heme-Fe and favors iron influx and efflux in intestinal cells. Iron 100-104 heme oxygenase 1 Homo sapiens 40-56 21622902-0 2011 Nrf-2 regulates cyclosporine-stimulated HO-1 expression in gingiva. Cyclosporine 16-28 heme oxygenase 1 Homo sapiens 40-44 21622902-6 2011 ERK inhibition significantly decreased CsA-stimulated Nrf-2 nuclear translocation and HO-1 mRNA expression. Cyclosporine 39-42 heme oxygenase 1 Homo sapiens 86-90 21622902-1 2011 Cyclosporine-A (CsA) stimulates heme oxygenase-1 (HO-1) expression in the gingiva, but the regulation and the role of HO-1 in gingival overgrowth are not well-understood. Cyclosporine 0-14 heme oxygenase 1 Homo sapiens 32-48 21622902-8 2011 These findings suggest that CsA-stimulated HO-1 expression is mediated through the activation of ERK, and that Nrf-2 plays a protective role against CsA-induced gingival fibrosis by modulating collagen turnover-related genes. Cyclosporine 28-31 heme oxygenase 1 Homo sapiens 43-47 21622902-1 2011 Cyclosporine-A (CsA) stimulates heme oxygenase-1 (HO-1) expression in the gingiva, but the regulation and the role of HO-1 in gingival overgrowth are not well-understood. Cyclosporine 0-14 heme oxygenase 1 Homo sapiens 50-54 21622902-1 2011 Cyclosporine-A (CsA) stimulates heme oxygenase-1 (HO-1) expression in the gingiva, but the regulation and the role of HO-1 in gingival overgrowth are not well-understood. Cyclosporine 16-19 heme oxygenase 1 Homo sapiens 32-48 21622902-1 2011 Cyclosporine-A (CsA) stimulates heme oxygenase-1 (HO-1) expression in the gingiva, but the regulation and the role of HO-1 in gingival overgrowth are not well-understood. Cyclosporine 16-19 heme oxygenase 1 Homo sapiens 50-54 21622902-3 2011 The aim of this study was to examine the role of Nrf-2 in the regulation of CsA-stimulated HO-1 expression in human gingival fibroblasts. Cyclosporine 76-79 heme oxygenase 1 Homo sapiens 91-95 21622902-5 2011 Treatment with siNrf-2, but not with an NF-kappaB inhibitor, reduced CsA-stimulated HO-1 mRNA expression. Cyclosporine 69-72 heme oxygenase 1 Homo sapiens 84-88 21994478-4 2011 Treatment with cilostazol increased HO-1 expression and phosphorylation of AMPK in a dose- and time-dependent manner. Cilostazol 15-25 heme oxygenase 1 Homo sapiens 36-40 21994478-0 2011 Cilostazol Inhibits Vascular Smooth Muscle Cell Proliferation and Reactive Oxygen Species Production through Activation of AMP-activated Protein Kinase Induced by Heme Oxygenase-1. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 163-179 21372182-9 2011 TcdA increased HO-1 expression as seen by immunohistochemistry. tcda 0-4 heme oxygenase 1 Homo sapiens 15-19 21372182-10 2011 These results suggest that the HO-1/CO pathway exerts a protective role in TcdA-induced enteritis and that its pharmacological modulation might be important for the management of C. difficile-associated disease. tcda 75-79 heme oxygenase 1 Homo sapiens 31-35 21994478-5 2011 Cilostazol also significantly decreased platelet-derived growth factor (PDGF)-induced VSMC proliferation and ROS production by activating AMPK induced by HO-1. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 154-158 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Cilostazol 68-78 heme oxygenase 1 Homo sapiens 42-46 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Cilostazol 68-78 heme oxygenase 1 Homo sapiens 182-186 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Reactive Oxygen Species 124-127 heme oxygenase 1 Homo sapiens 42-46 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Cilostazol 163-173 heme oxygenase 1 Homo sapiens 42-46 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Cilostazol 163-173 heme oxygenase 1 Homo sapiens 182-186 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Reactive Oxygen Species 270-273 heme oxygenase 1 Homo sapiens 42-46 21994478-6 2011 Pharmacological and genetic inhibition of HO-1 and AMPK blocked the cilostazol-induced inhibition of cell proliferation and ROS production.These data suggest that cilostazol-induced HO-1 expression and AMPK activation might attenuate PDGF-induced VSMC proliferation and ROS production. Reactive Oxygen Species 270-273 heme oxygenase 1 Homo sapiens 182-186 21765607-6 2011 Taurine chloramine increased heme oxygenase-1 expression and heme oxygenase activity. N-chlorotaurine 0-18 heme oxygenase 1 Homo sapiens 29-45 21549697-6 2011 In contrast to the level of HO-1 expression in high tidal volume group, pretreatment with hemin, an inducer of HO-1, further up-regulated HO-1 expression. Hemin 90-95 heme oxygenase 1 Homo sapiens 28-32 21549697-6 2011 In contrast to the level of HO-1 expression in high tidal volume group, pretreatment with hemin, an inducer of HO-1, further up-regulated HO-1 expression. Hemin 90-95 heme oxygenase 1 Homo sapiens 111-115 21549697-6 2011 In contrast to the level of HO-1 expression in high tidal volume group, pretreatment with hemin, an inducer of HO-1, further up-regulated HO-1 expression. Hemin 90-95 heme oxygenase 1 Homo sapiens 111-115 21722625-6 2011 High dose of glucose enhanced the expression of HO-1 mRNA and HO-1 protein in a time-dependent manner. Glucose 13-20 heme oxygenase 1 Homo sapiens 48-52 21722625-6 2011 High dose of glucose enhanced the expression of HO-1 mRNA and HO-1 protein in a time-dependent manner. Glucose 13-20 heme oxygenase 1 Homo sapiens 62-66 21722625-7 2011 LAB enhanced the expression of HO-1 mRNA and HO-1 protein in a dose-dependent manner treated with high dose of glucose. Glucose 111-118 heme oxygenase 1 Homo sapiens 31-35 21722625-7 2011 LAB enhanced the expression of HO-1 mRNA and HO-1 protein in a dose-dependent manner treated with high dose of glucose. Glucose 111-118 heme oxygenase 1 Homo sapiens 45-49 21689633-4 2011 Hyperoside further enhanced the cellular antioxidant defense system through increasing the activity of heme oxygenase-1 (HO-1), and by up-regulating HO-1 expression. hyperoside 0-10 heme oxygenase 1 Homo sapiens 103-119 21689633-4 2011 Hyperoside further enhanced the cellular antioxidant defense system through increasing the activity of heme oxygenase-1 (HO-1), and by up-regulating HO-1 expression. hyperoside 0-10 heme oxygenase 1 Homo sapiens 121-125 21689633-4 2011 Hyperoside further enhanced the cellular antioxidant defense system through increasing the activity of heme oxygenase-1 (HO-1), and by up-regulating HO-1 expression. hyperoside 0-10 heme oxygenase 1 Homo sapiens 149-153 21689633-6 2011 Collectively, our data provide the first description of the mechanism underlying hyperoside"s ability to attenuate H(2)O(2)-induced cell damage, namely this compound interacts with the MAPK-dependent Keap(1)-Nrf(2)-ARE signaling pathway to up-regulate HO-1 expression and enhance intracellular antioxidant activity. hyperoside 81-91 heme oxygenase 1 Homo sapiens 252-256 21752292-8 2011 Although most oil components may reduce oxidant stress by undergoing reduction, orange oil was demonstrated to have the ability to induce long-lasting gene expression of several antioxidant enzymes linked to Nrf2, including HO-1, NQO1, GCLm and GCLc, and to mitigate the pro-inflammatory signaling of endotoxin in cell culture systems. orange oil 80-90 heme oxygenase 1 Homo sapiens 224-228 21752292-10 2011 Treatment with the aerosolized oil preparation increased baseline levels of nasal mucosal HO-1 expression in 9 of 12 subjects. Oils 31-34 heme oxygenase 1 Homo sapiens 90-94 21793851-6 2011 Carbon monoxide (CO), a reaction product of heme oxygenase-1 (HO-1), reduces oxidative stress and inflammatory response and protects cells from ER stress. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 44-60 21793851-6 2011 Carbon monoxide (CO), a reaction product of heme oxygenase-1 (HO-1), reduces oxidative stress and inflammatory response and protects cells from ER stress. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 62-66 21793851-6 2011 Carbon monoxide (CO), a reaction product of heme oxygenase-1 (HO-1), reduces oxidative stress and inflammatory response and protects cells from ER stress. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 44-60 21793851-6 2011 Carbon monoxide (CO), a reaction product of heme oxygenase-1 (HO-1), reduces oxidative stress and inflammatory response and protects cells from ER stress. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 62-66 21793851-8 2011 In addition to its anti-inflammatory effects and antiapoptotic effects, HO-1 plays an important role in insulin release and glucose metabolism. Glucose 124-131 heme oxygenase 1 Homo sapiens 72-76 21787730-6 2011 Levels of NQO1 and HO1 mRNA are also increased by -fold and -fold following 24h exposure to 5 and 10 muM cadmium. Cadmium 105-112 heme oxygenase 1 Homo sapiens 19-22 21765607-9 2011 Combined, these results indicate that the taurine chloramine, produced and released endogenously by the activated neutrophils, can protect the macrophages in inflamed tissues from the H(2)O(2)-derived apoptosis not only by increasing the expression of cytoprotective enzymes like heme oxygenase-1 and catalase, but also by increasing the intracellular antioxidant GSH level. Taurine 42-49 heme oxygenase 1 Homo sapiens 280-309 21765607-9 2011 Combined, these results indicate that the taurine chloramine, produced and released endogenously by the activated neutrophils, can protect the macrophages in inflamed tissues from the H(2)O(2)-derived apoptosis not only by increasing the expression of cytoprotective enzymes like heme oxygenase-1 and catalase, but also by increasing the intracellular antioxidant GSH level. chloramine 50-60 heme oxygenase 1 Homo sapiens 280-309 21765607-7 2011 The taurine chloramine-derived upregulation of heme oxygenase-1 expression was blocked by inhibition of ERK phosphorylation. Taurine 4-11 heme oxygenase 1 Homo sapiens 47-63 21412259-0 2011 Heme oxygenase-1 protects human melanocytes from H2O2-induced oxidative stress via the Nrf2-ARE pathway. Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 0-16 21412259-6 2011 Our study demonstrated that heme oxygenase-1 (HO-1) was the most induced antioxidant gene in PIG1 cells after treatment with H(2)O(2). Water 125-130 heme oxygenase 1 Homo sapiens 28-44 21765607-7 2011 The taurine chloramine-derived upregulation of heme oxygenase-1 expression was blocked by inhibition of ERK phosphorylation. chloramine 12-22 heme oxygenase 1 Homo sapiens 47-63 21412259-6 2011 Our study demonstrated that heme oxygenase-1 (HO-1) was the most induced antioxidant gene in PIG1 cells after treatment with H(2)O(2). Water 125-130 heme oxygenase 1 Homo sapiens 46-50 21412259-8 2011 In addition, the H(2)O(2)-induced Nrf2-ARE/HO-1 pathway was confirmed in primary cultured human melanocytes by examining the expression and translocation of Nrf2 and HO-1. Hydrogen Peroxide 17-25 heme oxygenase 1 Homo sapiens 43-47 21327520-0 2011 Ferrearin C induces apoptosis via heme oxygenase-1 (HO-1) induction in neuroblastoma. ferrearin c 0-11 heme oxygenase 1 Homo sapiens 34-50 21412259-8 2011 In addition, the H(2)O(2)-induced Nrf2-ARE/HO-1 pathway was confirmed in primary cultured human melanocytes by examining the expression and translocation of Nrf2 and HO-1. Hydrogen Peroxide 17-25 heme oxygenase 1 Homo sapiens 166-170 21412259-9 2011 These data suggested that regulation of the Nrf2/HO-1 pathway can reduce H(2)O(2)-induced oxidative damage in human melanocytes. Hydrogen Peroxide 73-81 heme oxygenase 1 Homo sapiens 49-53 21327520-0 2011 Ferrearin C induces apoptosis via heme oxygenase-1 (HO-1) induction in neuroblastoma. ferrearin c 0-11 heme oxygenase 1 Homo sapiens 52-56 21861350-5 2011 Moreover, HO-1 mRNA expression were also significantly induced by sodium arsenite exposure, and a definite dose-effect relationship was confirmed (P < 0.01). sodium arsenite 66-81 heme oxygenase 1 Homo sapiens 10-14 21593686-0 2011 Propofol upregulates heme oxygenase-1 through activation of ERKs in human umbilical vein endothelial cells under oxidative stress conditions. Propofol 0-8 heme oxygenase 1 Homo sapiens 21-37 21593686-2 2011 We examined the effect of propofol on the regulation of HO-1 expression and its activity in human umbilical vein endothelial cells (HUVECs) under oxidative stress conditions. Propofol 26-34 heme oxygenase 1 Homo sapiens 56-60 21593686-3 2011 We further assessed whether extracellular signal-regulated kinases (ERKs), cJun-N-terminal kinases (JNKs), and p38-mitogen-activated protein kinase mediate propofol-induced HO-1 expression. Propofol 156-164 heme oxygenase 1 Homo sapiens 173-177 21593686-8 2011 HO-1 activity was determined in microsomal fractions from HUVECs by monitoring the conversion of heme into bilirubin. Heme 97-101 heme oxygenase 1 Homo sapiens 0-4 21593686-8 2011 HO-1 activity was determined in microsomal fractions from HUVECs by monitoring the conversion of heme into bilirubin. Bilirubin 107-116 heme oxygenase 1 Homo sapiens 0-4 21593686-11 2011 RESULTS: Under oxidative stress conditions, HO-1 expression and activity were increased by propofol in a dose-dependent and time-dependent manner. Propofol 91-99 heme oxygenase 1 Homo sapiens 44-48 21593686-12 2011 PD98059, but not SB203580 or SP600125, effectively reduced propofol-induced HO-1 protein levels. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 heme oxygenase 1 Homo sapiens 76-80 21593686-12 2011 PD98059, but not SB203580 or SP600125, effectively reduced propofol-induced HO-1 protein levels. Propofol 59-67 heme oxygenase 1 Homo sapiens 76-80 21593686-15 2011 CONCLUSIONS: These findings show that, under oxidative stress conditions, propofol induces HO-1 expression in HUVECs and this effect is mediated, at least in part, via ERKs pathways. Propofol 74-82 heme oxygenase 1 Homo sapiens 91-95 21481475-6 2011 HMOX1, an indicator of stress due to reactive oxygen intermediates (ROIs) and/or metals, was upregulated at the later time point as well. reactive oxygen 37-52 heme oxygenase 1 Homo sapiens 0-5 21861350-6 2011 CONCLUSION: No elevation of Nrf2 transcription was found, while inorganic arsenic could increase the Nrf2-regulated NQO1 and HO-1 mRNA expression in human hepatocytes. Arsenic 74-81 heme oxygenase 1 Homo sapiens 125-129 21367956-4 2011 Treatment of mouse epidermal JB6 cells with zerumbone caused a marked increase of Nrf2 nuclear translocation followed by the promoter activity of HO-1, and also enhanced direct binding of Nrf2 to the antioxidant response element. zerumbone 44-53 heme oxygenase 1 Homo sapiens 146-150 21620381-0 2011 MG132 enhances neurite outgrowth in neurons overexpressing mutant TAR DNA-binding protein-43 via increase of HO-1. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 heme oxygenase 1 Homo sapiens 109-113 21620381-5 2011 Heme oxygenase-1 (HO-1) known as antioxidase was restored by MG132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 61-66 heme oxygenase 1 Homo sapiens 0-16 21620381-5 2011 Heme oxygenase-1 (HO-1) known as antioxidase was restored by MG132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 61-66 heme oxygenase 1 Homo sapiens 18-22 21620381-8 2011 However, MG132 increased the expression of HO-1 independent of the Nrf2 pathway. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 9-14 heme oxygenase 1 Homo sapiens 43-47 21367956-5 2011 Moreover, knockdown of Nrf2 in JB6 cells diminished the zerumbone-induced upregulation of HO-1. zerumbone 56-65 heme oxygenase 1 Homo sapiens 90-94 21367956-8 2011 Interestingly, when JB6 cells were treated with carbon monoxide-releasing molecule that mimics the HO-1 activity, the TPA-induced ROS production was markedly reduced. Carbon Monoxide 48-63 heme oxygenase 1 Homo sapiens 99-103 21367956-8 2011 Interestingly, when JB6 cells were treated with carbon monoxide-releasing molecule that mimics the HO-1 activity, the TPA-induced ROS production was markedly reduced. Tetradecanoylphorbol Acetate 118-121 heme oxygenase 1 Homo sapiens 99-103 21367956-8 2011 Interestingly, when JB6 cells were treated with carbon monoxide-releasing molecule that mimics the HO-1 activity, the TPA-induced ROS production was markedly reduced. Reactive Oxygen Species 130-133 heme oxygenase 1 Homo sapiens 99-103 21396424-6 2011 Furthermore, [6]-gingerol treatment up-regulated the mRNA and protein expression of antioxidant enzymes such as gamma-glutamylcysteine ligase (GCL) and heme oxygenase-1 (HO-1), the rate limiting enzymes in the glutathione biosynthesis and the degradation of heme, respectively. gingerol 17-25 heme oxygenase 1 Homo sapiens 152-168 21426408-6 2011 Heme oxygenase encoding genes were subsequently cloned to study consequences of either gene product on cell viability, demonstrating that HO-1 but not HO-2 overexpression offers protection from stress-induced cell death in MTT assays. monooxyethylene trimethylolpropane tristearate 223-226 heme oxygenase 1 Homo sapiens 138-142 21671166-4 2011 The results showed that CdCl(2) and nano-TiO(2) at a low concentration of 0.75 total toxic unit (TU) exerted an additive effects on HO-1 gene expression, CAT activities and MDA concentrations. cdcl 24-28 heme oxygenase 1 Homo sapiens 132-136 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Heme 93-97 heme oxygenase 1 Homo sapiens 0-16 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Heme 93-97 heme oxygenase 1 Homo sapiens 18-22 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Iron 124-136 heme oxygenase 1 Homo sapiens 0-16 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Iron 124-136 heme oxygenase 1 Homo sapiens 18-22 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 138-153 heme oxygenase 1 Homo sapiens 0-16 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 138-153 heme oxygenase 1 Homo sapiens 18-22 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 155-157 heme oxygenase 1 Homo sapiens 0-16 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 155-157 heme oxygenase 1 Homo sapiens 18-22 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Biliverdine 164-174 heme oxygenase 1 Homo sapiens 0-16 21622183-1 2011 Heme oxygenase-1 (HO-1) catalyzes the first and rate-limiting step in the metabolism of free heme into equimolar amounts of ferrous iron, carbon monoxide (CO), and biliverdin. Biliverdine 164-174 heme oxygenase 1 Homo sapiens 18-22 21622183-4 2011 HO-1 represses inflammation by removing the pro-inflammatory molecule heme and by generating CO and the bile pigments, biliverdin and bilirubin. Heme 70-74 heme oxygenase 1 Homo sapiens 0-4 21622183-4 2011 HO-1 represses inflammation by removing the pro-inflammatory molecule heme and by generating CO and the bile pigments, biliverdin and bilirubin. Biliverdine 119-129 heme oxygenase 1 Homo sapiens 0-4 21622183-4 2011 HO-1 represses inflammation by removing the pro-inflammatory molecule heme and by generating CO and the bile pigments, biliverdin and bilirubin. Bilirubin 134-143 heme oxygenase 1 Homo sapiens 0-4 21136273-10 2011 However, blockade of HO-1 using ZnPPIX totally abolished BMP-6-regulated MMP-9 activation in MCF-7 cells. zinc protoporphyrin 32-38 heme oxygenase 1 Homo sapiens 21-25 21333731-8 2011 On the other hand, RSG notably increased the expression of key antioxidant transcription factor Nrf2 and antioxidant enzyme HO-1 in a PPARgamma-dependent manner, leading to the elimination of excessive ROS. Reactive Oxygen Species 202-205 heme oxygenase 1 Homo sapiens 124-128 21333731-10 2011 Furthermore, the activation of Akt and MAPKs was involved in the effect of RSG on Nrf2, HO-1 and COX-2. Rosiglitazone 75-78 heme oxygenase 1 Homo sapiens 88-92 21693314-2 2011 MATERIALS AND METHODS: To increase the expression of HO-1, both donors and recipients were injected with heme through the abdomen before the operation. Heme 105-109 heme oxygenase 1 Homo sapiens 53-57 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 54-60 heme oxygenase 1 Homo sapiens 100-116 21781496-0 2011 [Effects of HO-1 gene expression on proliferation of imatinib resistant CML cells]. Imatinib Mesylate 53-61 heme oxygenase 1 Homo sapiens 12-16 21781496-1 2011 OBJECTIVE: To investigate the effect of heme oxygenase-1 (HO-1) expression on cell growth and apoptosis in imatinib resistant chronic myeloid leukemia (CML) cells (K562/A02-IM), and explore the relationship between HO-1 gene and CML. Imatinib Mesylate 107-115 heme oxygenase 1 Homo sapiens 40-56 21781496-1 2011 OBJECTIVE: To investigate the effect of heme oxygenase-1 (HO-1) expression on cell growth and apoptosis in imatinib resistant chronic myeloid leukemia (CML) cells (K562/A02-IM), and explore the relationship between HO-1 gene and CML. Imatinib Mesylate 107-115 heme oxygenase 1 Homo sapiens 58-62 21781496-3 2011 Different concentrations of hemin were used to induce HO-1 expression of K562/A02-IM, HO-1 expression at different time was detected by RT-PCR and Western blot analysis. Hemin 28-33 heme oxygenase 1 Homo sapiens 54-58 21781496-3 2011 Different concentrations of hemin were used to induce HO-1 expression of K562/A02-IM, HO-1 expression at different time was detected by RT-PCR and Western blot analysis. Hemin 28-33 heme oxygenase 1 Homo sapiens 86-90 21781496-9 2011 When HO-1 was inhibited by ZPP, the cells survival was sharply decreased compared to that of the control group (P < 0.05). zinc protoporphyrin 27-30 heme oxygenase 1 Homo sapiens 5-9 21443188-0 2011 tBHQ-induced HO-1 expression is mediated by calcium through regulation of Nrf2 binding to enhancer and polymerase II to promoter region of HO-1. 2-tert-butylhydroquinone 0-4 heme oxygenase 1 Homo sapiens 13-17 21443188-0 2011 tBHQ-induced HO-1 expression is mediated by calcium through regulation of Nrf2 binding to enhancer and polymerase II to promoter region of HO-1. 2-tert-butylhydroquinone 0-4 heme oxygenase 1 Homo sapiens 139-143 21443188-0 2011 tBHQ-induced HO-1 expression is mediated by calcium through regulation of Nrf2 binding to enhancer and polymerase II to promoter region of HO-1. Calcium 44-51 heme oxygenase 1 Homo sapiens 13-17 21443188-0 2011 tBHQ-induced HO-1 expression is mediated by calcium through regulation of Nrf2 binding to enhancer and polymerase II to promoter region of HO-1. Calcium 44-51 heme oxygenase 1 Homo sapiens 139-143 21443188-2 2011 The goal of this study was to examine the role of calcium [Ca(2+)] in regulating a well-known phenolic chemopreventive compound tertiary-butylhydroquinone (tBHQ) activation of Nrf2 and induction of Nrf2 downstream target gene heme-oxygenase (HO-1). Calcium 50-57 heme oxygenase 1 Homo sapiens 242-246 21443188-2 2011 The goal of this study was to examine the role of calcium [Ca(2+)] in regulating a well-known phenolic chemopreventive compound tertiary-butylhydroquinone (tBHQ) activation of Nrf2 and induction of Nrf2 downstream target gene heme-oxygenase (HO-1). 2-Butylhydroquinone 137-154 heme oxygenase 1 Homo sapiens 242-246 21443188-2 2011 The goal of this study was to examine the role of calcium [Ca(2+)] in regulating a well-known phenolic chemopreventive compound tertiary-butylhydroquinone (tBHQ) activation of Nrf2 and induction of Nrf2 downstream target gene heme-oxygenase (HO-1). 2-tert-butylhydroquinone 156-160 heme oxygenase 1 Homo sapiens 242-246 21443188-3 2011 tBHQ alone caused Nrf2 nuclear localization and induced HO-1 mRNA and protein expression in a dose-dependent manner. 2-tert-butylhydroquinone 0-4 heme oxygenase 1 Homo sapiens 56-60 21443188-4 2011 Using RT-PCR and Western blotting, we showed that tBHQ-induced transcription of HO-1 is Ca(2+)-dependent. 2-tert-butylhydroquinone 50-54 heme oxygenase 1 Homo sapiens 80-84 21443188-5 2011 Chelation of [Ca(2+)](ext) or [Ca(2+)](intra) by EGTA or BAPTA attenuated tBHQ-induced HO-1. Egtazic Acid 49-53 heme oxygenase 1 Homo sapiens 87-91 21443188-5 2011 Chelation of [Ca(2+)](ext) or [Ca(2+)](intra) by EGTA or BAPTA attenuated tBHQ-induced HO-1. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 57-62 heme oxygenase 1 Homo sapiens 87-91 21443188-5 2011 Chelation of [Ca(2+)](ext) or [Ca(2+)](intra) by EGTA or BAPTA attenuated tBHQ-induced HO-1. 2-tert-butylhydroquinone 74-78 heme oxygenase 1 Homo sapiens 87-91 21443188-8 2011 Additionally, EGTA and BAPTA treatments decreased basal nuclear phosphorylation of CREB and decreased tBHQ-induced Nrf2-CBP binding and Nrf2 binding to enhancer as well as polymerase II binding to the promoter of HO-1 gene. Egtazic Acid 14-18 heme oxygenase 1 Homo sapiens 213-217 21443188-8 2011 Additionally, EGTA and BAPTA treatments decreased basal nuclear phosphorylation of CREB and decreased tBHQ-induced Nrf2-CBP binding and Nrf2 binding to enhancer as well as polymerase II binding to the promoter of HO-1 gene. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 23-28 heme oxygenase 1 Homo sapiens 213-217 21443188-8 2011 Additionally, EGTA and BAPTA treatments decreased basal nuclear phosphorylation of CREB and decreased tBHQ-induced Nrf2-CBP binding and Nrf2 binding to enhancer as well as polymerase II binding to the promoter of HO-1 gene. 2-tert-butylhydroquinone 102-106 heme oxygenase 1 Homo sapiens 213-217 21443188-9 2011 Furthermore, tBHQ in combination with higher [Ca(2+)](ext) augmented HO-1 induction both in vitro and in vivo, indicating that the modulation of [Ca(2+)](int) could be used as an adjuvant to increase the efficacy of chemopreventive agents. 2-tert-butylhydroquinone 13-17 heme oxygenase 1 Homo sapiens 69-73 21443188-10 2011 Taken together, our results indicated that in addition to tBHQ-induced oxidative stress-mediated Nrf2 translocation, HO-1 induction by tBHQ also appears to be dependent on a series of Ca(2+)-regulated mechanisms. 2-tert-butylhydroquinone 135-139 heme oxygenase 1 Homo sapiens 117-121 21518986-4 2011 METHODS AND RESULTS: We generated bitransgenic mice that overexpress human heme oxygenase-1 under doxycycline control in an inducible, lung-specific manner. Doxycycline 98-109 heme oxygenase 1 Homo sapiens 75-91 21572963-4 2011 Western blot analysis revealed that the expression of the cytoprotective enzyme, heme oxygenase-1 (HO-1), was induced and coincident with the anti-inflammatory action of NaHS. sodium bisulfide 170-174 heme oxygenase 1 Homo sapiens 81-97 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 54-60 heme oxygenase 1 Homo sapiens 118-122 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 54-60 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 54-60 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 54-60 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 128-134 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 128-134 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. Oxidopamine 128-134 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 244-263 heme oxygenase 1 Homo sapiens 100-116 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 244-263 heme oxygenase 1 Homo sapiens 118-122 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 244-263 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 244-263 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 244-263 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 265-269 heme oxygenase 1 Homo sapiens 100-116 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 265-269 heme oxygenase 1 Homo sapiens 118-122 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 265-269 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 265-269 heme oxygenase 1 Homo sapiens 190-194 21397656-7 2011 In another experiment, exposure of C6 glioma cells to 6-OHDA resulted in an increased expression of heme oxygenase-1 (HO-1) and 6-OHDA-induced cytotoxicity was effectively suppressed by the HO-1 inducer SnCl(2) and aggravated by HO-1 inhibitor zinc protoporphyrin (ZnPP), supporting the cytoprotective role of HO-1. zinc protoporphyrin 265-269 heme oxygenase 1 Homo sapiens 190-194 21397656-8 2011 To elucidate the molecular mechanism underlying 6-OHDA-mediated HO-1 induction, we have examined the possible involvement of NF-E2-related factor 2 (Nrf2), which plays an important role in the transcriptional regulation of phase II detoxifying and antioxidant enzymes. Oxidopamine 48-54 heme oxygenase 1 Homo sapiens 64-68 21397656-10 2011 Taken together these findings suggest that HO-1 up-regulation via Nrf2 activation may mediate the cellular adaptive survival response to 6-OHDA-induced nitrosative cell death in C6 glioma cells. Oxidopamine 137-143 heme oxygenase 1 Homo sapiens 43-47 21248240-3 2011 An additional feature of neuronal oxidative stress is the up-regulation of the inducible enzyme heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Biliverdine 156-166 heme oxygenase 1 Homo sapiens 96-112 21354100-3 2011 The present study aimed at clarifying whether exposure of endothelial cells to pro-inflammatory HOCl-HDL impacts on expression of heme oxygenase-1, a potential rescue pathway against endothelial dysfunction. Hypochlorous Acid 96-100 heme oxygenase 1 Homo sapiens 130-146 21354100-4 2011 Our findings revealed that HDL modified by HOCl, added as reagent or generated enzymatically, induced phosphorylation of p42/44 mitogen-activated protein kinase, expression of transcription factor early growth response-1 (Egr-1) and enhanced expression of heme oxygenase-1 in human endothelial cells. Hypochlorous Acid 43-47 heme oxygenase 1 Homo sapiens 256-272 21248240-3 2011 An additional feature of neuronal oxidative stress is the up-regulation of the inducible enzyme heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). ammonium ferrous sulfate 168-174 heme oxygenase 1 Homo sapiens 96-112 21248240-3 2011 An additional feature of neuronal oxidative stress is the up-regulation of the inducible enzyme heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 180-195 heme oxygenase 1 Homo sapiens 96-112 21248240-3 2011 An additional feature of neuronal oxidative stress is the up-regulation of the inducible enzyme heme oxygenase-1 (HO-1), which catabolizes heme to generate biliverdin, Fe(2+), and carbon monoxide (CO). Carbon Monoxide 197-199 heme oxygenase 1 Homo sapiens 96-112 21414301-0 2011 Celastrol induces expression of heme oxygenase-1 through ROS/Nrf2/ARE signaling in the HaCaT cells. celastrol 0-9 heme oxygenase 1 Homo sapiens 32-48 21399873-8 2011 Microarray data from cells treated with nifurtimox and TM validated the induction of oxidative stress, as many Nrf2 target genes (HMOX1, GCLM, SLC7A11 and SRXN1) (p<10(-5)) were upregulated. Nifurtimox 40-50 heme oxygenase 1 Homo sapiens 130-135 21529713-5 2011 Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 56-60 21529713-5 2011 Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 56-60 21529713-5 2011 Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Heme 37-41 heme oxygenase 1 Homo sapiens 56-60 21529713-5 2011 Carbon monoxide (CO), a byproduct of heme catabolism by HO-1, prevents further accumulation of circulating free heme after Plasmodium infection, suppressing the pathogenesis of ECM. Heme 112-116 heme oxygenase 1 Homo sapiens 56-60 21787712-11 2011 Furthermore, the indirect effect of Cd through HO-1 (genetic polymorphism and prevalence of long GT(n) repeats) and 20-HETE was inconclusive. Cadmium 36-38 heme oxygenase 1 Homo sapiens 47-51 21315065-0 2011 Lipoic acid ameliorates arsenic trioxide-induced HO-1 expression and oxidative stress in THP-1 monocytes and macrophages. Thioctic Acid 0-11 heme oxygenase 1 Homo sapiens 49-53 21315065-0 2011 Lipoic acid ameliorates arsenic trioxide-induced HO-1 expression and oxidative stress in THP-1 monocytes and macrophages. Arsenic Trioxide 24-40 heme oxygenase 1 Homo sapiens 49-53 21315065-3 2011 We found that a non-lethal concentration of arsenic trioxide (1 muM) significantly induced the expression of heme oxygenase-1 (HO-1), a response biomarker to arsenic, without stimulating measurable superoxide production. Arsenic Trioxide 44-60 heme oxygenase 1 Homo sapiens 109-125 21315065-3 2011 We found that a non-lethal concentration of arsenic trioxide (1 muM) significantly induced the expression of heme oxygenase-1 (HO-1), a response biomarker to arsenic, without stimulating measurable superoxide production. Arsenic Trioxide 44-60 heme oxygenase 1 Homo sapiens 127-131 21315065-3 2011 We found that a non-lethal concentration of arsenic trioxide (1 muM) significantly induced the expression of heme oxygenase-1 (HO-1), a response biomarker to arsenic, without stimulating measurable superoxide production. Arsenic 44-51 heme oxygenase 1 Homo sapiens 109-125 21315065-3 2011 We found that a non-lethal concentration of arsenic trioxide (1 muM) significantly induced the expression of heme oxygenase-1 (HO-1), a response biomarker to arsenic, without stimulating measurable superoxide production. Arsenic 44-51 heme oxygenase 1 Homo sapiens 127-131 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. zinc protoporphyrin 58-77 heme oxygenase 1 Homo sapiens 31-35 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. zinc protoporphyrin 58-77 heme oxygenase 1 Homo sapiens 143-147 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. zinc protoporphyrin 79-83 heme oxygenase 1 Homo sapiens 31-35 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. zinc protoporphyrin 79-83 heme oxygenase 1 Homo sapiens 143-147 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. Arsenic 97-104 heme oxygenase 1 Homo sapiens 31-35 21315065-4 2011 Co-treatment of cells with the HO-1 competitive inhibitor zinc protoporphyrin (Znpp) potentiated arsenic-induced cytotoxicity, indicating that HO-1 confers a cytoprotective effect against arsenic toxicity. Arsenic 97-104 heme oxygenase 1 Homo sapiens 143-147 21414301-0 2011 Celastrol induces expression of heme oxygenase-1 through ROS/Nrf2/ARE signaling in the HaCaT cells. ros 57-60 heme oxygenase 1 Homo sapiens 32-48 21414301-1 2011 We previously demonstrated that celastrol, a quinone methide triterpenoid derived from the medicinal plant Tripterygium wilfordii, exerts its anti-inflammatory activity through up-regulation of heme oxygenase-1 (HO-1) expression in the keratinocytes. celastrol 32-41 heme oxygenase 1 Homo sapiens 194-210 21414301-1 2011 We previously demonstrated that celastrol, a quinone methide triterpenoid derived from the medicinal plant Tripterygium wilfordii, exerts its anti-inflammatory activity through up-regulation of heme oxygenase-1 (HO-1) expression in the keratinocytes. celastrol 32-41 heme oxygenase 1 Homo sapiens 212-216 21414301-2 2011 In this study, we examined the signaling pathways that lead to the up-regulation of HO-1 expression by celastrol. celastrol 103-112 heme oxygenase 1 Homo sapiens 84-88 21414301-3 2011 In HaCaT cells, celastrol-induced HO-1 expression was dependent on ROS generation. celastrol 16-25 heme oxygenase 1 Homo sapiens 34-38 21414301-3 2011 In HaCaT cells, celastrol-induced HO-1 expression was dependent on ROS generation. ros 67-70 heme oxygenase 1 Homo sapiens 34-38 21414301-4 2011 ERK and p38 MAPK were major MAPK pathways responsible for celastrol-induced HO-1 expression. celastrol 58-67 heme oxygenase 1 Homo sapiens 76-80 21414301-6 2011 Nrf2 knockdown using small interfering RNA (siRNA) inhibited celastrol-induced HO-1 expression. celastrol 61-70 heme oxygenase 1 Homo sapiens 79-83 21414301-9 2011 Taken together, our results indicate that celastrol can activate the ROS-ERK/p38-Nrf2-ARE signaling cascades leading to the up-regulation of HO-1 which is partly responsible for its anti-inflammatory activity in the keratinocytes. celastrol 42-51 heme oxygenase 1 Homo sapiens 141-145 21414301-9 2011 Taken together, our results indicate that celastrol can activate the ROS-ERK/p38-Nrf2-ARE signaling cascades leading to the up-regulation of HO-1 which is partly responsible for its anti-inflammatory activity in the keratinocytes. ros 69-72 heme oxygenase 1 Homo sapiens 141-145 21382015-0 2011 The anti-inflammatory effects of dimethyl fumarate in astrocytes involve glutathione and haem oxygenase-1. Dimethyl Fumarate 33-50 heme oxygenase 1 Homo sapiens 89-105 21382015-2 2011 DMF modifies glutathione (GSH) levels that can induce expression of the anti-inflammatory protein HO-1 (haem oxygenase-1). Dimethyl Fumarate 0-3 heme oxygenase 1 Homo sapiens 98-120 21382015-2 2011 DMF modifies glutathione (GSH) levels that can induce expression of the anti-inflammatory protein HO-1 (haem oxygenase-1). Glutathione 13-24 heme oxygenase 1 Homo sapiens 98-120 21382015-2 2011 DMF modifies glutathione (GSH) levels that can induce expression of the anti-inflammatory protein HO-1 (haem oxygenase-1). Glutathione 26-29 heme oxygenase 1 Homo sapiens 98-120 21382015-10 2011 In astrocytes, BG-12 increased HO-1 mRNA levels and effects on nitrite levels were blocked by an HO-1 inhibitor. Nitrites 63-70 heme oxygenase 1 Homo sapiens 97-101 21483629-7 2011 Bach1 and HMOX-1 RNA were also downregulated by LS treatment (P < 0.01), while Nrf2 protein was increased (P < 0.05). leucylserine 48-50 heme oxygenase 1 Homo sapiens 10-16 21325398-5 2011 Insulin induced HOX-1 expression in a time- and dose-dependent manner, an effect attenuated by selective inhibitors of ERK1/2 (PD98059), Src (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine), and c-Jun terminal kinases 1 and 2 (SP600125) pathways. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 127-134 heme oxygenase 1 Homo sapiens 16-21 21544718-7 2011 Heme oxygenase-1 is considered to be an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron and biliverdin, while SIRT1 is the mammalian homologue of the yeast silent information regulator (Sir)-2, which are involved in the suppression of inflammatory mediators or factors that may be used to improve atopy-related symptoms. Heme 76-80 heme oxygenase 1 Homo sapiens 0-16 21544718-7 2011 Heme oxygenase-1 is considered to be an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron and biliverdin, while SIRT1 is the mammalian homologue of the yeast silent information regulator (Sir)-2, which are involved in the suppression of inflammatory mediators or factors that may be used to improve atopy-related symptoms. Carbon Monoxide 84-99 heme oxygenase 1 Homo sapiens 0-16 21544718-7 2011 Heme oxygenase-1 is considered to be an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron and biliverdin, while SIRT1 is the mammalian homologue of the yeast silent information regulator (Sir)-2, which are involved in the suppression of inflammatory mediators or factors that may be used to improve atopy-related symptoms. Iron 106-110 heme oxygenase 1 Homo sapiens 0-16 21544718-7 2011 Heme oxygenase-1 is considered to be an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron and biliverdin, while SIRT1 is the mammalian homologue of the yeast silent information regulator (Sir)-2, which are involved in the suppression of inflammatory mediators or factors that may be used to improve atopy-related symptoms. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 0-16 21168266-6 2011 Celastrol"s ability to modulate the expression of pro-inflammatory cytokines, MHC II, HO-1, iNOS, NF-kappaB, Notch-1, AKT/mTOR, CXCR4, TRAIL receptors DR4 and DR5, CHOP, JNK, VEGF, adhesion molecules, proteasome activity, topoisomerase II, potassium channels, and heat shock response has been reported. celastrol 0-9 heme oxygenase 1 Homo sapiens 86-90 21325398-5 2011 Insulin induced HOX-1 expression in a time- and dose-dependent manner, an effect attenuated by selective inhibitors of ERK1/2 (PD98059), Src (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine), and c-Jun terminal kinases 1 and 2 (SP600125) pathways. AG 1879 142-206 heme oxygenase 1 Homo sapiens 16-21 21325398-5 2011 Insulin induced HOX-1 expression in a time- and dose-dependent manner, an effect attenuated by selective inhibitors of ERK1/2 (PD98059), Src (4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolo[3,4-d] pyrimidine), and c-Jun terminal kinases 1 and 2 (SP600125) pathways. pyrazolanthrone 245-253 heme oxygenase 1 Homo sapiens 16-21 21198350-0 2011 Association of (GT)n repeats promoter polymorphism of heme oxygenase-1 gene with serum bilirubin levels in healthy Indian adults. Bilirubin 87-96 heme oxygenase 1 Homo sapiens 54-70 21198350-1 2011 AIM: The present study was undertaken to investigate a length polymorphism of (GT)n repeats of the heme oxygenase-1 (HMOX-1) gene and its association with serum bilirubin levels in apparently healthy adults. Bilirubin 161-170 heme oxygenase 1 Homo sapiens 99-115 21198350-1 2011 AIM: The present study was undertaken to investigate a length polymorphism of (GT)n repeats of the heme oxygenase-1 (HMOX-1) gene and its association with serum bilirubin levels in apparently healthy adults. Bilirubin 161-170 heme oxygenase 1 Homo sapiens 117-123 21238556-0 2011 Upregulation of heme oxygenase-1 as an adaptive mechanism for protection against crotonaldehyde in human umbilical vein endothelial cells. 2-butenal 81-95 heme oxygenase 1 Homo sapiens 16-32 21462044-5 2011 HO-1 is an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron, and biliverdin, all of which are involved in the suppression of inflammatory mediators. Heme 47-51 heme oxygenase 1 Homo sapiens 0-4 21462044-5 2011 HO-1 is an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron, and biliverdin, all of which are involved in the suppression of inflammatory mediators. Carbon Monoxide 55-70 heme oxygenase 1 Homo sapiens 0-4 21462044-5 2011 HO-1 is an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron, and biliverdin, all of which are involved in the suppression of inflammatory mediators. Iron 77-81 heme oxygenase 1 Homo sapiens 0-4 21462044-5 2011 HO-1 is an antioxidant enzyme that catabolizes heme to carbon monoxide, free iron, and biliverdin, all of which are involved in the suppression of inflammatory mediators. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 0-4 21660147-6 2011 Moreover, cilostazol increased HO-1 protein and mRNA expression. Cilostazol 10-20 heme oxygenase 1 Homo sapiens 31-35 21660147-7 2011 Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). Cilostazol 0-10 heme oxygenase 1 Homo sapiens 19-23 21660147-7 2011 Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). Zinc protoporphyrin 70-74 heme oxygenase 1 Homo sapiens 19-23 21660147-7 2011 Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). copper protoporphyrin IX 87-111 heme oxygenase 1 Homo sapiens 19-23 21660147-7 2011 Cilostazol-induced HO-1 induction was markedly attenuated not only by ZnPP but also by copper-protoporphyrin IX (CuPP). cupp 113-117 heme oxygenase 1 Homo sapiens 19-23 21660147-8 2011 In an assay measuring peroxisome proliferator-activated receptor-gamma (PPAR-gamma) transcription activity, cilostazol directly increased PPAR-gamma transcriptional activity which was completely abolished by HO-1 inhibitor. Cilostazol 108-118 heme oxygenase 1 Homo sapiens 208-212 21660147-9 2011 Furthermore, increased PPAR-gamma activity by cilostazol and rosiglitazone was completely abolished in cells transfected with HO-1 siRNA. Cilostazol 46-56 heme oxygenase 1 Homo sapiens 126-130 21660147-9 2011 Furthermore, increased PPAR-gamma activity by cilostazol and rosiglitazone was completely abolished in cells transfected with HO-1 siRNA. Rosiglitazone 61-74 heme oxygenase 1 Homo sapiens 126-130 21660147-10 2011 Taken together, these results indicate that cilostazol up-regulates HO-1 and protects cells against TNF-alpha-induced endothelial cytotoxicity via a PPAR-gamma-dependent pathway. Cilostazol 44-54 heme oxygenase 1 Homo sapiens 68-72 21146245-7 2011 Indeed, in CYP2E1-expressing HepG2 cells exposed to ethanol, the expression of ISR target genes (HMOX-1, GCLC, AsnS, IGFBP-1, GADD34,CHOP, ATF3, CHAC1) was induced. Ethanol 52-59 heme oxygenase 1 Homo sapiens 97-103 21660147-0 2011 HO-1 Induced by Cilostazol Protects Against TNF-alpha-associated Cytotoxicity via a PPAR-gamma-dependent Pathway in Human Endothelial Cells. Cilostazol 16-26 heme oxygenase 1 Homo sapiens 0-4 21660147-2 2011 In our previous study, cilostazol was found to increase the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in synovial cells. Cilostazol 23-33 heme oxygenase 1 Homo sapiens 97-113 21660147-2 2011 In our previous study, cilostazol was found to increase the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) in synovial cells. Cilostazol 23-33 heme oxygenase 1 Homo sapiens 115-119 21660147-3 2011 Thus, the present study was undertaken to examine whether cilostazol is able to counteract tumor necrosis factor-alpha (TNF-alpha)-induced cell death in endothelial cells via the induction of HO-1 expression. Cilostazol 58-68 heme oxygenase 1 Homo sapiens 192-196 21660147-5 2011 Pretreatment with cilostazol markedly reduced TNF-alpha-induced viability loss in the HUVECs, which was reversed by zinc protoporphyrine IX (ZnPP), an inhibitor of HO-1. Cilostazol 18-28 heme oxygenase 1 Homo sapiens 164-168 21660147-5 2011 Pretreatment with cilostazol markedly reduced TNF-alpha-induced viability loss in the HUVECs, which was reversed by zinc protoporphyrine IX (ZnPP), an inhibitor of HO-1. zinc protoporphyrine ix 116-139 heme oxygenase 1 Homo sapiens 164-168 21660147-5 2011 Pretreatment with cilostazol markedly reduced TNF-alpha-induced viability loss in the HUVECs, which was reversed by zinc protoporphyrine IX (ZnPP), an inhibitor of HO-1. Zinc protoporphyrin 141-145 heme oxygenase 1 Homo sapiens 164-168 21172416-7 2011 Transcriptomics, western blot and immunofluorescence showed an induction of both HO-1 and NQO1 with Cd and Diq exposure, but not with CsA treatment. Cadmium 100-102 heme oxygenase 1 Homo sapiens 81-85 21172416-7 2011 Transcriptomics, western blot and immunofluorescence showed an induction of both HO-1 and NQO1 with Cd and Diq exposure, but not with CsA treatment. Diquat 107-110 heme oxygenase 1 Homo sapiens 81-85 21172416-9 2011 siRNA knock down of HO-1, but not NQO1, enhanced Cd induced H(2)O(2) production and Cd induced toxicity. Cadmium 49-51 heme oxygenase 1 Homo sapiens 20-24 21172416-9 2011 siRNA knock down of HO-1, but not NQO1, enhanced Cd induced H(2)O(2) production and Cd induced toxicity. Hydrogen Peroxide 60-68 heme oxygenase 1 Homo sapiens 20-24 21172416-9 2011 siRNA knock down of HO-1, but not NQO1, enhanced Cd induced H(2)O(2) production and Cd induced toxicity. Cadmium 84-86 heme oxygenase 1 Homo sapiens 20-24 21481317-3 2011 The effect of low dose propofol on expression of heme oxygenase-1 (HO-1) was confirmed by Real-time quantitative PCR. Propofol 23-31 heme oxygenase 1 Homo sapiens 49-65 21481317-3 2011 The effect of low dose propofol on expression of heme oxygenase-1 (HO-1) was confirmed by Real-time quantitative PCR. Propofol 23-31 heme oxygenase 1 Homo sapiens 67-71 21481317-5 2011 And low dose propofol increased the expression of HO-1 mRNA in a dose-dependment manner. Propofol 13-21 heme oxygenase 1 Homo sapiens 50-54 21481317-6 2011 CONCLUSION: Low dose propofol affects the biological behavior of esophageal squamous cell carcinoma cell line Eca109, which has a relationship with increasing the expression of HO-1. Propofol 21-29 heme oxygenase 1 Homo sapiens 177-181 21352808-4 2011 Notably, GW501516 up-regulated the expression of antioxidant genes, such as glutathione peroxidase 1, thioredoxin 1, manganese superoxide dismutase and heme oxygenase 1. GW 501516 9-17 heme oxygenase 1 Homo sapiens 152-168 21361338-3 2011 Most of the TCEs having two different kinds of nonenolizable cyano enones in rings A and C are highly potent suppressors of induction of inducible nitric oxide synthase stimulated with interferon-gamma and are highly potent inducers of the cytoprotective enzymes heme oxygenase-1 and NAD(P)H:quinone oxidoreductase-1. tces 12-16 heme oxygenase 1 Homo sapiens 263-316 21361338-3 2011 Most of the TCEs having two different kinds of nonenolizable cyano enones in rings A and C are highly potent suppressors of induction of inducible nitric oxide synthase stimulated with interferon-gamma and are highly potent inducers of the cytoprotective enzymes heme oxygenase-1 and NAD(P)H:quinone oxidoreductase-1. cyano enones 61-73 heme oxygenase 1 Homo sapiens 263-316 21238556-5 2011 In this study, we examined the effects of crotonaldehyde on HO-1 induction and determined the signaling pathways in human umbilical vein endothelial cells (HUVECs). 2-butenal 42-56 heme oxygenase 1 Homo sapiens 60-64 21238556-6 2011 Inhibition of the protein kinase C-delta (PKC-delta) and p38 pathways resulted in significant blockage of crotonaldehyde-mediated HO-1 induction. 2-butenal 106-120 heme oxygenase 1 Homo sapiens 130-134 21238556-7 2011 Crotonaldehyde treatment caused a dramatic increase in translocation of NF-E2 related factor (Nrf2), leading to induction of HO-1. 2-butenal 0-14 heme oxygenase 1 Homo sapiens 125-129 21238556-8 2011 In addition, small interfering RNA knockdown of Nrf2 and treatment with the specific HO-1 inhibitor ZnPP exhibited an obvious increase of apoptosis of crotonaldehyde-treated HUVECs. zinc protoporphyrin 100-104 heme oxygenase 1 Homo sapiens 85-89 21238556-8 2011 In addition, small interfering RNA knockdown of Nrf2 and treatment with the specific HO-1 inhibitor ZnPP exhibited an obvious increase of apoptosis of crotonaldehyde-treated HUVECs. 2-butenal 151-165 heme oxygenase 1 Homo sapiens 85-89 21238556-9 2011 Taken together, our results demonstrated that crotonaldehyde-induced HO-1 expression is mediated by the PKC-delta-p38 MAPK-Nrf2-HO-1 pathway in HUVECs, which is an adaptive response to oxidative stress. 2-butenal 46-60 heme oxygenase 1 Homo sapiens 69-73 21238556-9 2011 Taken together, our results demonstrated that crotonaldehyde-induced HO-1 expression is mediated by the PKC-delta-p38 MAPK-Nrf2-HO-1 pathway in HUVECs, which is an adaptive response to oxidative stress. 2-butenal 46-60 heme oxygenase 1 Homo sapiens 128-132 21185934-3 2011 Under hypoxia, the expression of major iron homeostasis genes including transferrin, transferrin receptor, ceruloplasmin, and heme oxygenase-1 is activated by hypoxia-inducible factors to provide increased iron availability for erythropoiesis in an attempt to enhance oxygen uptake and delivery to hypoxic cells. Iron 39-43 heme oxygenase 1 Homo sapiens 126-142 21185934-3 2011 Under hypoxia, the expression of major iron homeostasis genes including transferrin, transferrin receptor, ceruloplasmin, and heme oxygenase-1 is activated by hypoxia-inducible factors to provide increased iron availability for erythropoiesis in an attempt to enhance oxygen uptake and delivery to hypoxic cells. Iron 206-210 heme oxygenase 1 Homo sapiens 126-142 21374664-7 2011 IL-10 and heme oxygenase-1 (HO-1) were up-regulated in patients receiving norfloxacin and correlated with norfloxacin in a concentration-dependent manner, whereas proinflammatory inducible nitric oxide synthase, cyclooxygenase-2, and nuclear factor-kappaB behaved inversely. Norfloxacin 74-85 heme oxygenase 1 Homo sapiens 10-26 21307647-0 2011 Lipopolysaccaride induces autophagic signaling in macrophages via a TLR4, heme oxygenase-1 dependent pathway. lipopolysaccaride 0-17 heme oxygenase 1 Homo sapiens 74-90 21374664-7 2011 IL-10 and heme oxygenase-1 (HO-1) were up-regulated in patients receiving norfloxacin and correlated with norfloxacin in a concentration-dependent manner, whereas proinflammatory inducible nitric oxide synthase, cyclooxygenase-2, and nuclear factor-kappaB behaved inversely. Norfloxacin 74-85 heme oxygenase 1 Homo sapiens 28-32 21374664-7 2011 IL-10 and heme oxygenase-1 (HO-1) were up-regulated in patients receiving norfloxacin and correlated with norfloxacin in a concentration-dependent manner, whereas proinflammatory inducible nitric oxide synthase, cyclooxygenase-2, and nuclear factor-kappaB behaved inversely. Norfloxacin 106-117 heme oxygenase 1 Homo sapiens 10-26 21374664-7 2011 IL-10 and heme oxygenase-1 (HO-1) were up-regulated in patients receiving norfloxacin and correlated with norfloxacin in a concentration-dependent manner, whereas proinflammatory inducible nitric oxide synthase, cyclooxygenase-2, and nuclear factor-kappaB behaved inversely. Norfloxacin 106-117 heme oxygenase 1 Homo sapiens 28-32 20836698-8 2011 Lentiviral transduction with the VECAD-HO-1 construct attenuated the increase in blood pressure (p < 0.05), improved vascular relaxation, increased plasma adiponectin, and prevented the elevation in urinary protein and plasma creatinine in Ang II-treated rats. Creatinine 229-239 heme oxygenase 1 Homo sapiens 39-43 21374664-10 2011 Neutrophilic in vitro assays showed that the effect of LPS on proinflammatory mediator levels in the presence of norfloxacin was abrogated by significantly increasing IL-10 and HO-1 expression. Norfloxacin 113-124 heme oxygenase 1 Homo sapiens 177-181 21374664-13 2011 CONCLUSION: Although the described association does not necessarily mean causality, an IL-10-mediated HO-1-induced anti-inflammatory mechanism is present in patients with cirrhosis receiving norfloxacin, that is directly associated with cell-modulating events in these patients. Norfloxacin 191-202 heme oxygenase 1 Homo sapiens 102-106 21259326-4 2011 In this study, we demonstrate that MGF24-induced heme oxygenase-1 up-regulation is dependent on activation of protein kinase Cepsilon and NF-E2-related factor-2 (Nrf2). mgf24 35-40 heme oxygenase 1 Homo sapiens 49-65 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Heme 73-77 heme oxygenase 1 Homo sapiens 0-16 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Heme 73-77 heme oxygenase 1 Homo sapiens 18-22 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Biliverdine 105-115 heme oxygenase 1 Homo sapiens 0-16 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Biliverdine 105-115 heme oxygenase 1 Homo sapiens 18-22 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Iron 122-126 heme oxygenase 1 Homo sapiens 0-16 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Iron 122-126 heme oxygenase 1 Homo sapiens 18-22 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 0-16 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 18-22 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Carbon Monoxide 148-150 heme oxygenase 1 Homo sapiens 0-16 21373265-1 2011 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme, followed by production of biliverdin, free iron and carbon monoxide (CO). Carbon Monoxide 148-150 heme oxygenase 1 Homo sapiens 18-22 21373265-6 2011 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid, and dextran sulfate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the gastrointestinal tract. Indomethacin 155-167 heme oxygenase 1 Homo sapiens 45-49 21373265-6 2011 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid, and dextran sulfate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the gastrointestinal tract. Trinitrobenzenesulfonic Acid 207-236 heme oxygenase 1 Homo sapiens 45-49 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 18-22 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 0-16 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 18-22 20563825-3 2011 Astroglial induction of the Hmox1 gene by beta-amyloid, pro-inflammatory cytokines and hydrogen peroxide promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergy failure amply documented in AD-affected neural tissues. Hydrogen Peroxide 87-104 heme oxygenase 1 Homo sapiens 28-33 20563825-3 2011 Astroglial induction of the Hmox1 gene by beta-amyloid, pro-inflammatory cytokines and hydrogen peroxide promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergy failure amply documented in AD-affected neural tissues. Iron 161-165 heme oxygenase 1 Homo sapiens 28-33 20563825-3 2011 Astroglial induction of the Hmox1 gene by beta-amyloid, pro-inflammatory cytokines and hydrogen peroxide promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergy failure amply documented in AD-affected neural tissues. Iron 232-236 heme oxygenase 1 Homo sapiens 28-33 20563825-4 2011 Glial HO-1 expression may also impact cell survival and neuroplasticity in AD by modulating brain sterol/oxysterol metabolism and the degradation of tau by the proteasome. Sterols 98-104 heme oxygenase 1 Homo sapiens 6-10 20563825-4 2011 Glial HO-1 expression may also impact cell survival and neuroplasticity in AD by modulating brain sterol/oxysterol metabolism and the degradation of tau by the proteasome. Oxysterols 105-114 heme oxygenase 1 Homo sapiens 6-10 20563825-5 2011 Suppression of glial HO-1 activity by pharmacological or other means may confer neuroprotection in AD by curtailing iron-mediated neurotoxicity. Iron 116-120 heme oxygenase 1 Homo sapiens 21-25 21062351-9 2011 The capacity of melatonin to modulate Nrf2 pathway was associated with increased heme oxygenase-1 (HO-1) expression, which strengthens antioxidant defense. Melatonin 16-25 heme oxygenase 1 Homo sapiens 81-97 21373265-6 2011 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid, and dextran sulfate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the gastrointestinal tract. Dextran Sulfate 242-264 heme oxygenase 1 Homo sapiens 45-49 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Heme 78-82 heme oxygenase 1 Homo sapiens 0-16 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Heme 78-82 heme oxygenase 1 Homo sapiens 18-22 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. ferrous iron 86-98 heme oxygenase 1 Homo sapiens 0-16 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. ferrous iron 86-98 heme oxygenase 1 Homo sapiens 18-22 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Carbon Monoxide 100-115 heme oxygenase 1 Homo sapiens 0-16 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Carbon Monoxide 100-115 heme oxygenase 1 Homo sapiens 18-22 20563825-1 2011 Heme oxygenase-1 (HO-1), a 32 kDa stress protein mediating the degradation of heme to ferrous iron, carbon monoxide and biliverdin/bilirubin, has been implicated in the pathogenesis of Alzheimer disease (AD) and other aging-related neurodegenerative disorders. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 0-16 21073519-0 2011 Melatonin synergistically increases resveratrol-induced heme oxygenase-1 expression through the inhibition of ubiquitin-dependent proteasome pathway: a possible role in neuroprotection. Melatonin 0-9 heme oxygenase 1 Homo sapiens 56-72 21073519-0 2011 Melatonin synergistically increases resveratrol-induced heme oxygenase-1 expression through the inhibition of ubiquitin-dependent proteasome pathway: a possible role in neuroprotection. Resveratrol 36-47 heme oxygenase 1 Homo sapiens 56-72 21073519-4 2011 We assessed whether the co-administration of melatonin and RSV shows synergistic effects in terms of their neuroprotective properties through HO-1. Melatonin 45-54 heme oxygenase 1 Homo sapiens 142-146 21073519-4 2011 We assessed whether the co-administration of melatonin and RSV shows synergistic effects in terms of their neuroprotective properties through HO-1. Resveratrol 59-62 heme oxygenase 1 Homo sapiens 142-146 21073519-5 2011 RSV significantly increased the expression levels of HO-1 protein in a concentration-dependent manner both in primary cortical neurons and in astrocytes, while melatonin per se did not. Resveratrol 0-3 heme oxygenase 1 Homo sapiens 53-57 21073519-6 2011 Melatonin + RSV showed a synergistic increase in the expression levels of HO-1 protein but not in the HO-1 mRNA level compared to either melatonin or RSV alone, which is mediated by the activation of PI3K-Akt pathway. Melatonin 0-9 heme oxygenase 1 Homo sapiens 74-78 21073519-6 2011 Melatonin + RSV showed a synergistic increase in the expression levels of HO-1 protein but not in the HO-1 mRNA level compared to either melatonin or RSV alone, which is mediated by the activation of PI3K-Akt pathway. Resveratrol 12-15 heme oxygenase 1 Homo sapiens 74-78 21073519-6 2011 Melatonin + RSV showed a synergistic increase in the expression levels of HO-1 protein but not in the HO-1 mRNA level compared to either melatonin or RSV alone, which is mediated by the activation of PI3K-Akt pathway. Resveratrol 12-15 heme oxygenase 1 Homo sapiens 102-106 21259326-5 2011 MGF24 induces nuclear translocation of Nrf2, and siRNA knockdown of Nrf2 or of heme oxygenase-1 prevents MGF24-induced heme oxygenase-1 up-regulation and neuroprotection of SH-SY5Y cells against 6-hydroxydopamine-induced cell death. Oxidopamine 195-212 heme oxygenase 1 Homo sapiens 79-95 21073519-8 2011 The blockade of HO-1 induction by shRNA attenuated HO-1 induction by melatonin + RSV and hindered the neuroprotective effects against oxidative stress induced by H(2) O(2) . Melatonin 69-78 heme oxygenase 1 Homo sapiens 16-20 21073519-8 2011 The blockade of HO-1 induction by shRNA attenuated HO-1 induction by melatonin + RSV and hindered the neuroprotective effects against oxidative stress induced by H(2) O(2) . Melatonin 69-78 heme oxygenase 1 Homo sapiens 51-55 21073519-8 2011 The blockade of HO-1 induction by shRNA attenuated HO-1 induction by melatonin + RSV and hindered the neuroprotective effects against oxidative stress induced by H(2) O(2) . Resveratrol 81-84 heme oxygenase 1 Homo sapiens 16-20 21073519-8 2011 The blockade of HO-1 induction by shRNA attenuated HO-1 induction by melatonin + RSV and hindered the neuroprotective effects against oxidative stress induced by H(2) O(2) . Resveratrol 81-84 heme oxygenase 1 Homo sapiens 51-55 21073519-9 2011 The treatment of MG132 + RSV mimicked the effects of melatonin + RSV, and melatonin + RSV inhibited ubiquitination of HO-1. Melatonin 74-83 heme oxygenase 1 Homo sapiens 118-122 21073519-10 2011 These data suggest that melatonin potentiates the neuroprotective effect of RSV against oxidative injury, by enhancing HO-1 induction through inhibiting ubiquitination-dependent proteasome pathway, which may provide an effective means to treat neurodegenerative disorders. Melatonin 24-33 heme oxygenase 1 Homo sapiens 119-123 21073519-10 2011 These data suggest that melatonin potentiates the neuroprotective effect of RSV against oxidative injury, by enhancing HO-1 induction through inhibiting ubiquitination-dependent proteasome pathway, which may provide an effective means to treat neurodegenerative disorders. Resveratrol 76-79 heme oxygenase 1 Homo sapiens 119-123 21259326-5 2011 MGF24 induces nuclear translocation of Nrf2, and siRNA knockdown of Nrf2 or of heme oxygenase-1 prevents MGF24-induced heme oxygenase-1 up-regulation and neuroprotection of SH-SY5Y cells against 6-hydroxydopamine-induced cell death. Oxidopamine 195-212 heme oxygenase 1 Homo sapiens 119-135 21259326-9 2011 Taken together, these results demonstrate that PKC activity is needed for MGF24"s activation of Nrf2, which in turn increases heme oxygenase-1 expression, a critical event in mediating MGF24"s neuroprotection against 6-hydroxydopamine-induced apoptosis. Oxidopamine 217-234 heme oxygenase 1 Homo sapiens 126-142 21206978-6 2011 In addition, we administered an HO-1 inhibitor, zinc protoporphyrin IX, to mice with subcutaneous T24 tumors to assess the modulation of angiogenesis in solid tumors in vivo. zinc protoporphyrin 48-70 heme oxygenase 1 Homo sapiens 32-36 20938987-0 2011 Curcumin induces heme oxygenase-1 in normal human skin fibroblasts through redox signaling: relevance for anti-aging intervention. Curcumin 0-8 heme oxygenase 1 Homo sapiens 17-33 20938987-2 2011 METHODS AND RESULTS: Early passage young human skin fibroblasts treated with low doses of curcumin (below 20 muM) showed a time- and concentration-dependent induction of heme oxygenase-1 (HO-1), followed by compensatory increase in glutathione-S-transferase activity, GSH levels and GSH/GSSG ratio. Curcumin 90-98 heme oxygenase 1 Homo sapiens 170-186 20938987-2 2011 METHODS AND RESULTS: Early passage young human skin fibroblasts treated with low doses of curcumin (below 20 muM) showed a time- and concentration-dependent induction of heme oxygenase-1 (HO-1), followed by compensatory increase in glutathione-S-transferase activity, GSH levels and GSH/GSSG ratio. Curcumin 90-98 heme oxygenase 1 Homo sapiens 188-192 20938987-5 2011 The use of the antioxidant N-acetyl cysteine prevented the induction of HO-1 by curcumin. Acetylcysteine 27-44 heme oxygenase 1 Homo sapiens 72-76 20938987-5 2011 The use of the antioxidant N-acetyl cysteine prevented the induction of HO-1 by curcumin. Curcumin 80-88 heme oxygenase 1 Homo sapiens 72-76 20938987-6 2011 Pharmacological inhibition of phosphatidylinositol 3-kinase, but not other kinases, significantly prevented curcumin-induced HO-1 levels, which was corroborated by the induction of phospho-Akt levels by curcumin. Curcumin 108-116 heme oxygenase 1 Homo sapiens 125-129 20938987-7 2011 Late passage senescent cells already had higher HO-1 levels, and further induction of HO-1 by curcumin was considerably impaired. Curcumin 94-102 heme oxygenase 1 Homo sapiens 86-90 21281968-8 2011 NaAs systematically and significantly up-regulated a set of 35 genes, including several immune and stress genes, such as IL13, granulocyte-macrophage colony stimulating factor, lymphotoxin alpha and heme oxygenase-1 (HO-1). naas 0-4 heme oxygenase 1 Homo sapiens 199-215 21281968-8 2011 NaAs systematically and significantly up-regulated a set of 35 genes, including several immune and stress genes, such as IL13, granulocyte-macrophage colony stimulating factor, lymphotoxin alpha and heme oxygenase-1 (HO-1). naas 0-4 heme oxygenase 1 Homo sapiens 217-221 21281968-9 2011 Up-regulation of HO-1, a stress and immunosuppressive protein, was rapidly detectable, both in T cells and in PBMC treated with NaAs. naas 128-132 heme oxygenase 1 Homo sapiens 17-21 21281968-11 2011 Our findings demonstrate that, at least in vitro, inorganic arsenic acts directly on human T cells and impairs their activity, probably independently of HO-1 expression and monocyte-related accessory signals. Arsenic 60-67 heme oxygenase 1 Homo sapiens 153-157 21206978-9 2011 Hypoxic conditions induced upregulation of HO-1, VEGF, HIF-1alpha, HIF-2alpha, and COX-2 in T24 cells and increased VEGF secretion, which could be suppressed by zinc protoporphyrin IX. zinc protoporphyrin 161-183 heme oxygenase 1 Homo sapiens 43-47 21319354-8 2011 The HO-1 expression in the PF group was significantly higher than that in MF group at the corresponding time points (p < 0.05, respectively). pyrazofurin 27-29 heme oxygenase 1 Homo sapiens 4-8 21362092-3 2011 Cobalt protoporphyrin induced expression of HO-1 protein with maximal levels attaining a plateau at 48 hours. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 44-48 21148661-2 2011 The aim of this study was to evaluate the role of HO-1 in the regulation of insulin resistance and glucose tolerance in women with polycystic ovary syndrome (PCOS). Glucose 99-106 heme oxygenase 1 Homo sapiens 50-54 21362092-5 2011 Similarly, expression of HO-1 by introduction of a recombinant plasmid also showed a resistance to deoxycholate-induced apoptosis. Deoxycholic Acid 99-111 heme oxygenase 1 Homo sapiens 25-29 21144877-0 2011 Induction of apoptosis in human leukemia cells through the production of reactive oxygen species and activation of HMOX1 and Noxa by benzene, toluene, and o-xylene. Benzene 133-140 heme oxygenase 1 Homo sapiens 115-120 21144877-0 2011 Induction of apoptosis in human leukemia cells through the production of reactive oxygen species and activation of HMOX1 and Noxa by benzene, toluene, and o-xylene. Toluene 142-149 heme oxygenase 1 Homo sapiens 115-120 21144877-0 2011 Induction of apoptosis in human leukemia cells through the production of reactive oxygen species and activation of HMOX1 and Noxa by benzene, toluene, and o-xylene. 2-xylene 155-163 heme oxygenase 1 Homo sapiens 115-120 21106538-1 2011 Heme oxygenase-1 (HO-1) enzyme plays a critical role in metabolizing the excess heme generated during hemolysis. Heme 80-84 heme oxygenase 1 Homo sapiens 0-16 21106538-1 2011 Heme oxygenase-1 (HO-1) enzyme plays a critical role in metabolizing the excess heme generated during hemolysis. Heme 80-84 heme oxygenase 1 Homo sapiens 18-22 21144877-7 2011 Real-time RT-PCR revealed increased HMOX1 and Noxa expression in BZ-, TOL-, and o-XY-treated HL-60 cells, confirming the results of previous microarray analyses. Benzene 65-67 heme oxygenase 1 Homo sapiens 36-41 21144877-7 2011 Real-time RT-PCR revealed increased HMOX1 and Noxa expression in BZ-, TOL-, and o-XY-treated HL-60 cells, confirming the results of previous microarray analyses. 2-xylene 80-84 heme oxygenase 1 Homo sapiens 36-41 21148661-9 2011 Serum insulin and glucose levels and BMI showed a significant negative correlation with the level of HO-1 (P< 0.05). Glucose 18-25 heme oxygenase 1 Homo sapiens 101-105 20888844-8 2011 Sulforaphane stimulation for 4 h induced an Nrf2-dependent increase of Nqo1 and Hmox1 mRNA that remained elevated for 24 h, and the corresponding proteins remained elevated for over 48 h. In addition, peroxide-clearing activity and the levels of glutathione were elevated for more than 20 h after stimulation for 4 h with sulforaphane, resulting in an increased resistance to superoxide-induced cell damage. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 80-85 21148661-10 2011 CONCLUSIONS: Our results suggest that the HO-1-adiponectin axis may be associated with the regulation of insulin resistance and glucose intolerance in women with PCOS. Glucose 128-135 heme oxygenase 1 Homo sapiens 42-46 21099244-5 2011 Additionally, depletion of BLVRA reduced the H2O2-dependent induction of heme oxygenase-1 (HO-1) in young HDFs, but not in senescent cells, suggesting an aging-dependent differential modulation of responses to oxidative stress. Hydrogen Peroxide 45-49 heme oxygenase 1 Homo sapiens 73-89 21262447-5 2011 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 72-87 heme oxygenase 1 Homo sapiens 29-45 21262447-5 2011 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 72-87 heme oxygenase 1 Homo sapiens 47-51 21262447-5 2011 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 29-45 21262447-5 2011 An anti-inflammatory enzyme, heme oxygenase-1 (HO-1) and its metabolite carbon monoxide (CO), inhibit sFlt-1 and sEng release. Carbon Monoxide 89-91 heme oxygenase 1 Homo sapiens 47-51 21199573-0 2011 Ascorbic acid partly antagonizes resveratrol mediated heme oxygenase-1 but not paraoxonase-1 induction in cultured hepatocytes - role of the redox-regulated transcription factor Nrf2. Ascorbic Acid 0-13 heme oxygenase 1 Homo sapiens 54-70 21076374-4 2011 OBJECTIVE: To test whether preconditioning with fenoldopam (FD) induce HO-1 and protect kidneys against cold storage injury and whether HO-1 plays a role in protection. Fenoldopam 48-58 heme oxygenase 1 Homo sapiens 71-75 21081499-1 2011 Heme oxygenase-1 (HO-1) degrades heme and protects cells from oxidative challenge. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 21081499-2 2011 This antioxidant activity is thought to result from the HO-1 enzymatic activity, manifested by a decrease in the concentration of the pro-oxidant substrate heme, and an increase in the antioxidant product bilirubin. Heme 156-160 heme oxygenase 1 Homo sapiens 56-60 21081499-2 2011 This antioxidant activity is thought to result from the HO-1 enzymatic activity, manifested by a decrease in the concentration of the pro-oxidant substrate heme, and an increase in the antioxidant product bilirubin. Bilirubin 205-214 heme oxygenase 1 Homo sapiens 56-60 21171611-12 2011 The induction of oxidative stress and inflammatory responses (evaluated by HO-1 mRNA expression and TNF-alpha mRNA and protein expression) revealed a reduction in inflammogenicity upon iron loading and a more inflammogenic potency of DQ12 ascribed to undissociated SiOH interacting via H-bonding with cell membrane components. Iron 185-189 heme oxygenase 1 Homo sapiens 75-79 21199573-6 2011 Heme oxygenase-1 induction by resveratrol was accompanied by an increase in Nrf2 transactivation. Resveratrol 30-41 heme oxygenase 1 Homo sapiens 0-16 21199573-7 2011 Resveratrol mediated Nrf2 transactivation as well as heme oxygenase-1 induction were partly antagonized by 1000 mumol/l ascorbic acid. Ascorbic Acid 120-133 heme oxygenase 1 Homo sapiens 53-69 21199573-0 2011 Ascorbic acid partly antagonizes resveratrol mediated heme oxygenase-1 but not paraoxonase-1 induction in cultured hepatocytes - role of the redox-regulated transcription factor Nrf2. Resveratrol 33-44 heme oxygenase 1 Homo sapiens 54-70 21199573-9 2011 Addition of resveratrol to the cell culture medium produced relatively low levels of hydrogen peroxide which may be a positive hormetic redox-signal for Nrf2 dependent gene expression thereby driving heme oxygenase-1 induction. Resveratrol 12-23 heme oxygenase 1 Homo sapiens 200-216 21199573-3 2011 METHODS: The antioxidant enzymes heme oxygenase-1 and paraoxonase-1 were analysed for their mRNA and protein levels in HUH7 liver cells treated with 10 and 25 mumol/l resveratrol in the absence and presence of 100 and 1000 mumol/l ascorbic acid. Resveratrol 167-178 heme oxygenase 1 Homo sapiens 33-49 21199573-9 2011 Addition of resveratrol to the cell culture medium produced relatively low levels of hydrogen peroxide which may be a positive hormetic redox-signal for Nrf2 dependent gene expression thereby driving heme oxygenase-1 induction. Hydrogen Peroxide 85-102 heme oxygenase 1 Homo sapiens 200-216 21199573-5 2011 RESULTS: Here, we demonstrate that resveratrol induces the antioxidant enzymes heme oxygenase-1 and paraoxonase-1 in cultured hepatocytes. Resveratrol 35-46 heme oxygenase 1 Homo sapiens 79-95 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Heme 51-55 heme oxygenase 1 Homo sapiens 0-16 22162689-0 2011 Heme Oxygenase-1: A Critical Link between Iron Metabolism, Erythropoiesis, and Development. Iron 42-46 heme oxygenase 1 Homo sapiens 0-16 22162689-4 2011 Null mutation of Hmox1 results in significant embryonic mortality as well as anemia and defective iron recycling. Iron 98-102 heme oxygenase 1 Homo sapiens 17-22 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Heme 51-55 heme oxygenase 1 Homo sapiens 18-22 21905282-10 2011 SHXW essential oil suppressed Abeta-induced apoptosis and ROS production via an up-regulation of HO-1 and Nrf2 expression in SH-SY5Y cells. Oils, Volatile 5-18 heme oxygenase 1 Homo sapiens 97-101 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 0-16 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 18-22 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 0-16 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 18-22 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Iron 101-105 heme oxygenase 1 Homo sapiens 0-16 22254194-3 2011 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to generate carbon monoxide, biliverdin, and iron. Iron 101-105 heme oxygenase 1 Homo sapiens 18-22 21037234-4 2011 Induction of HO-1 by AICAR was blocked by the AMPK inhibitor compound C, the adenosine kinase inhibitor 5"-iodotubercidin, and by silencing AMPK-alpha(1/2) and was mimicked by the AMPK activator A-769662 and by infecting ECs with an adenovirus expressing constitutively active AMPK-alpha(1). 5-iodotubercidin 104-121 heme oxygenase 1 Homo sapiens 13-17 21037234-2 2011 Treatment of human ECs with the AMPK activator 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) stimulated a concentration- and time-dependent increase in HO-1 protein and mRNA expression that was associated with a prominent increase in nuclear factor-erythroid 2-related factor 2 (Nrf2) protein. acadesine 47-101 heme oxygenase 1 Homo sapiens 169-173 21037234-2 2011 Treatment of human ECs with the AMPK activator 5-aminoimidazole-4-carboxamide-1-beta-d-ribofuranoside (AICAR) stimulated a concentration- and time-dependent increase in HO-1 protein and mRNA expression that was associated with a prominent increase in nuclear factor-erythroid 2-related factor 2 (Nrf2) protein. acadesine 103-108 heme oxygenase 1 Homo sapiens 169-173 21037234-4 2011 Induction of HO-1 by AICAR was blocked by the AMPK inhibitor compound C, the adenosine kinase inhibitor 5"-iodotubercidin, and by silencing AMPK-alpha(1/2) and was mimicked by the AMPK activator A-769662 and by infecting ECs with an adenovirus expressing constitutively active AMPK-alpha(1). acadesine 21-26 heme oxygenase 1 Homo sapiens 13-17 20934533-0 2011 Cellular iron depletion weakens induction of heme oxygenase-1 by cadmium. Iron 9-13 heme oxygenase 1 Homo sapiens 45-61 21328623-0 2011 The outcome of 5-ALA-mediated photodynamic treatment in melanoma cells is influenced by vitamin C and heme oxygenase-1. Aminolevulinic Acid 15-20 heme oxygenase 1 Homo sapiens 102-118 22125774-2 2011 METHODS: SiO(2)-induced oxidative stress was assessed by examining formation of reactive oxygen species (ROS), the induction of superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), as well as cytotoxicity effect was evaluation by cell viability. Silicon Dioxide 9-15 heme oxygenase 1 Homo sapiens 159-175 22125774-2 2011 METHODS: SiO(2)-induced oxidative stress was assessed by examining formation of reactive oxygen species (ROS), the induction of superoxide dismutase (SOD) and heme oxygenase-1 (HO-1), as well as cytotoxicity effect was evaluation by cell viability. Silicon Dioxide 9-15 heme oxygenase 1 Homo sapiens 177-181 22125774-6 2011 The expression of SOD protein was not changed, whereas that of HO-1 was increased by SiO(2) nanoparticle exposure. silicon monoxide 85-88 heme oxygenase 1 Homo sapiens 63-67 22125774-8 2011 SiO(2) nanoparticles induce induction of HO-1 via Nrf-2-ERK MAP kinase pathway. Silicon Dioxide 0-6 heme oxygenase 1 Homo sapiens 41-45 20966033-1 2011 Heme oxygenase-1 (HO-1) induction by hemin or Panhematin protects against experimental pancreatitis. Hemin 37-42 heme oxygenase 1 Homo sapiens 0-16 20966033-1 2011 Heme oxygenase-1 (HO-1) induction by hemin or Panhematin protects against experimental pancreatitis. Hemin 37-42 heme oxygenase 1 Homo sapiens 18-22 20966033-1 2011 Heme oxygenase-1 (HO-1) induction by hemin or Panhematin protects against experimental pancreatitis. Hemin 46-56 heme oxygenase 1 Homo sapiens 0-16 20966033-1 2011 Heme oxygenase-1 (HO-1) induction by hemin or Panhematin protects against experimental pancreatitis. Hemin 46-56 heme oxygenase 1 Homo sapiens 18-22 20966033-2 2011 As a preclinical first step toward determining whether HO-1 upregulation is a viable target in acute pancreatitis (AP) patients, we tested the hypothesis that HO-1 expression in peripheral blood mononuclear cell (PBMC) subsets of hospitalized patients with mild AP is upregulated then normalizes upon recovery and that cells from AP patients have the potential to upregulate their HO-1 ex vivo if exposed to Panhematin. Hemin 408-418 heme oxygenase 1 Homo sapiens 159-163 20966033-2 2011 As a preclinical first step toward determining whether HO-1 upregulation is a viable target in acute pancreatitis (AP) patients, we tested the hypothesis that HO-1 expression in peripheral blood mononuclear cell (PBMC) subsets of hospitalized patients with mild AP is upregulated then normalizes upon recovery and that cells from AP patients have the potential to upregulate their HO-1 ex vivo if exposed to Panhematin. Hemin 408-418 heme oxygenase 1 Homo sapiens 159-163 20966033-6 2011 Panhematin induced HO-1 in ex vivo cultured AP PBMCs more readily than in HC or VC PBMCs. Hemin 0-10 heme oxygenase 1 Homo sapiens 19-23 21660298-4 2011 A PCR-based method was used to search for oxidized purine base products in selected 200 bp sequences in promoters and coding regions of the VEGF, TGF-beta1, HO-1, Egr1, and beta-actin genes while quantitative Southern blot analysis was used to detect oxidative damage to the mitochondrial genome in lung tissue from control subjects and COPD patients. purine 51-57 heme oxygenase 1 Homo sapiens 157-161 20934533-0 2011 Cellular iron depletion weakens induction of heme oxygenase-1 by cadmium. Cadmium 65-72 heme oxygenase 1 Homo sapiens 45-61 20934533-3 2011 In HCT-116 cells, cadmium increased heme oxygenase-1 enzymatic activity. Cadmium 18-25 heme oxygenase 1 Homo sapiens 36-52 20934533-4 2011 This effect of cadmium was weaker in cells made iron-deficient with the iron chelator, desferrioxamine, which was associated with repression of heme oxygenase-1 protein and mRNA expression. Cadmium 15-22 heme oxygenase 1 Homo sapiens 144-160 20934533-4 2011 This effect of cadmium was weaker in cells made iron-deficient with the iron chelator, desferrioxamine, which was associated with repression of heme oxygenase-1 protein and mRNA expression. Iron 48-52 heme oxygenase 1 Homo sapiens 144-160 20934533-4 2011 This effect of cadmium was weaker in cells made iron-deficient with the iron chelator, desferrioxamine, which was associated with repression of heme oxygenase-1 protein and mRNA expression. Iron 72-76 heme oxygenase 1 Homo sapiens 144-160 20934533-4 2011 This effect of cadmium was weaker in cells made iron-deficient with the iron chelator, desferrioxamine, which was associated with repression of heme oxygenase-1 protein and mRNA expression. Deferoxamine 87-102 heme oxygenase 1 Homo sapiens 144-160 20934533-5 2011 The repression by desferrioxamine of cadmium-induced heme oxygenase-1 upregulation was reversed upon iron replenishment of the cells. Deferoxamine 18-33 heme oxygenase 1 Homo sapiens 53-69 20934533-5 2011 The repression by desferrioxamine of cadmium-induced heme oxygenase-1 upregulation was reversed upon iron replenishment of the cells. Cadmium 37-44 heme oxygenase 1 Homo sapiens 53-69 20934533-5 2011 The repression by desferrioxamine of cadmium-induced heme oxygenase-1 upregulation was reversed upon iron replenishment of the cells. Iron 101-105 heme oxygenase 1 Homo sapiens 53-69 20934533-6 2011 Additionally, it was found that thiol antioxidants inhibited the heme oxygenase-1 upregulation caused by cadmium and also by ethacrynic acid, which each decreased intracellular glutathione as did buthionine sulfoxamine. Sulfhydryl Compounds 32-37 heme oxygenase 1 Homo sapiens 65-81 20934533-6 2011 Additionally, it was found that thiol antioxidants inhibited the heme oxygenase-1 upregulation caused by cadmium and also by ethacrynic acid, which each decreased intracellular glutathione as did buthionine sulfoxamine. Cadmium 105-112 heme oxygenase 1 Homo sapiens 65-81 20934533-6 2011 Additionally, it was found that thiol antioxidants inhibited the heme oxygenase-1 upregulation caused by cadmium and also by ethacrynic acid, which each decreased intracellular glutathione as did buthionine sulfoxamine. Ethacrynic Acid 125-140 heme oxygenase 1 Homo sapiens 65-81 20934533-6 2011 Additionally, it was found that thiol antioxidants inhibited the heme oxygenase-1 upregulation caused by cadmium and also by ethacrynic acid, which each decreased intracellular glutathione as did buthionine sulfoxamine. Glutathione 177-188 heme oxygenase 1 Homo sapiens 65-81 20934533-6 2011 Additionally, it was found that thiol antioxidants inhibited the heme oxygenase-1 upregulation caused by cadmium and also by ethacrynic acid, which each decreased intracellular glutathione as did buthionine sulfoxamine. Buthionine Sulfoximine 196-218 heme oxygenase 1 Homo sapiens 65-81 20934533-7 2011 Interestingly, cadmium and ethacrynic acid increased nuclear translocation of Nrf2 and subsequent heme oxygenase-1 expression, but buthionine sulfoxamine did not. Cadmium 15-22 heme oxygenase 1 Homo sapiens 98-114 20934533-7 2011 Interestingly, cadmium and ethacrynic acid increased nuclear translocation of Nrf2 and subsequent heme oxygenase-1 expression, but buthionine sulfoxamine did not. Ethacrynic Acid 27-42 heme oxygenase 1 Homo sapiens 98-114 20934533-8 2011 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin, and a superoxide scavenger (Tiron) inhibited cadmium-induced upregulation of heme oxygenase-1. diphenyleneiodonium 39-58 heme oxygenase 1 Homo sapiens 150-166 20934533-8 2011 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin, and a superoxide scavenger (Tiron) inhibited cadmium-induced upregulation of heme oxygenase-1. acetovanillone 63-71 heme oxygenase 1 Homo sapiens 150-166 20934533-8 2011 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin, and a superoxide scavenger (Tiron) inhibited cadmium-induced upregulation of heme oxygenase-1. Superoxides 79-89 heme oxygenase 1 Homo sapiens 150-166 20934533-8 2011 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin, and a superoxide scavenger (Tiron) inhibited cadmium-induced upregulation of heme oxygenase-1. 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt 101-106 heme oxygenase 1 Homo sapiens 150-166 20934533-8 2011 Furthermore, NADPH oxidase inhibitors (diphenyleneiodonium and apocynin, and a superoxide scavenger (Tiron) inhibited cadmium-induced upregulation of heme oxygenase-1. Cadmium 118-125 heme oxygenase 1 Homo sapiens 150-166 20934533-10 2011 It is concluded that adequate amounts of iron, which at the atomic level can serve as the pivotal element of heme in NADPH oxidase, must be present in cells to permit what appears to be thiol redox-sensitive, NADPH oxidase-dependent upregulation of heme oxygenase-1. Iron 41-45 heme oxygenase 1 Homo sapiens 249-265 20934533-10 2011 It is concluded that adequate amounts of iron, which at the atomic level can serve as the pivotal element of heme in NADPH oxidase, must be present in cells to permit what appears to be thiol redox-sensitive, NADPH oxidase-dependent upregulation of heme oxygenase-1. Sulfhydryl Compounds 186-191 heme oxygenase 1 Homo sapiens 249-265 20934533-11 2011 Thus, these findings are significant because they suggest that cells without adequate iron would be unable to fully express the stress gene, heme oxygenase-1, when confronted with the toxic metal, cadmium. Metals 190-195 heme oxygenase 1 Homo sapiens 141-157 20934533-11 2011 Thus, these findings are significant because they suggest that cells without adequate iron would be unable to fully express the stress gene, heme oxygenase-1, when confronted with the toxic metal, cadmium. Cadmium 197-204 heme oxygenase 1 Homo sapiens 141-157 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 21297920-4 2011 Under these circumstances, the cells exhibited global protein arginine methylation: this event was also reproduced by the cell treatment with hemin, a heme oxygenase-1 inducer. Arginine 62-70 heme oxygenase 1 Homo sapiens 151-167 21297920-4 2011 Under these circumstances, the cells exhibited global protein arginine methylation: this event was also reproduced by the cell treatment with hemin, a heme oxygenase-1 inducer. Hemin 142-147 heme oxygenase 1 Homo sapiens 151-167 21088618-1 2011 Heme oxygenase-1 (HO-1) is a stress-induced enzyme that catalyses the oxidation of heme to biliverdin. Heme 83-87 heme oxygenase 1 Homo sapiens 0-16 21088618-1 2011 Heme oxygenase-1 (HO-1) is a stress-induced enzyme that catalyses the oxidation of heme to biliverdin. Heme 83-87 heme oxygenase 1 Homo sapiens 18-22 21088618-1 2011 Heme oxygenase-1 (HO-1) is a stress-induced enzyme that catalyses the oxidation of heme to biliverdin. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 0-16 21088618-1 2011 Heme oxygenase-1 (HO-1) is a stress-induced enzyme that catalyses the oxidation of heme to biliverdin. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 18-22 20561893-3 2011 METHODS: A PowerWave XS plate reader was used to monitor the absorbance (as a function of time) of heme bound to purified truncated human heme oxygenase-1 (hHO-1) in the individual wells of a standard 96-well plate (with or without the addition of a test compound). Heme 99-103 heme oxygenase 1 Homo sapiens 138-154 20561893-3 2011 METHODS: A PowerWave XS plate reader was used to monitor the absorbance (as a function of time) of heme bound to purified truncated human heme oxygenase-1 (hHO-1) in the individual wells of a standard 96-well plate (with or without the addition of a test compound). Heme 99-103 heme oxygenase 1 Homo sapiens 156-161 20561893-4 2011 The degradation of heme by heme oxygenase-1 was initiated using l-ascorbic acid, and the collected relevant absorbance data were analyzed by three different methods to calculate the percent control activity occurring in wells containing test compounds relative to that occurring in control wells with no test compound present. Heme 19-23 heme oxygenase 1 Homo sapiens 27-43 20561893-4 2011 The degradation of heme by heme oxygenase-1 was initiated using l-ascorbic acid, and the collected relevant absorbance data were analyzed by three different methods to calculate the percent control activity occurring in wells containing test compounds relative to that occurring in control wells with no test compound present. Ascorbic Acid 64-79 heme oxygenase 1 Homo sapiens 27-43 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Heme 60-64 heme oxygenase 1 Homo sapiens 180-184 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 0-16 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 18-22 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Biliverdine 128-138 heme oxygenase 1 Homo sapiens 180-184 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Iron 167-171 heme oxygenase 1 Homo sapiens 0-16 20941616-1 2011 Heme oxygenase-1 (HO-1) is one of the three isoforms of the heme oxygenase enzyme that catabolyzes the degradation of heme into biliverdin with the production of free iron and CO. HO-1 is induced by its substrate and by other stimuli, including agents involved in oxidative stress and proinflammatory cytokines as well as several anti-inflammatory stimuli. Iron 167-171 heme oxygenase 1 Homo sapiens 18-22 21311138-6 2011 The mRNA and protein levels of ET-1 were decreased, and the mRNA and protein levels of HO-1 increased in the Danshensu groups in a dose-dependent manner compared with the high glucose group. Glucose 176-183 heme oxygenase 1 Homo sapiens 87-91 21850179-10 2011 HO-1, catalase, GST-pi, and Nrf-2 were elevated in ARPE-19 cells after treatment with different concentrations of canolol for 24 h. Finally, canolol was found to activate extracellular signal regulated kinase (ERK) phosphorylation in ARPE-19 cells under the condition, with or without t-BH. 4-vinyl-2,6-dimethoxyphenol 114-121 heme oxygenase 1 Homo sapiens 0-14 21850179-10 2011 HO-1, catalase, GST-pi, and Nrf-2 were elevated in ARPE-19 cells after treatment with different concentrations of canolol for 24 h. Finally, canolol was found to activate extracellular signal regulated kinase (ERK) phosphorylation in ARPE-19 cells under the condition, with or without t-BH. 4-vinyl-2,6-dimethoxyphenol 141-148 heme oxygenase 1 Homo sapiens 0-14 20974203-7 2011 We also found that Tg induced the expression of the antioxidant gene heme oxygenase-1 (HO-1) in a dose-dependent manner, whereas Tm had a weak effect on HO-1 induction. Thapsigargin 19-21 heme oxygenase 1 Homo sapiens 69-85 20974203-7 2011 We also found that Tg induced the expression of the antioxidant gene heme oxygenase-1 (HO-1) in a dose-dependent manner, whereas Tm had a weak effect on HO-1 induction. Thapsigargin 19-21 heme oxygenase 1 Homo sapiens 87-91 20974203-10 2011 Because actinomycin D inhibited HO-1 induction by Tg, the induction of HO-1 may be regulated at the transcriptional level. Dactinomycin 8-21 heme oxygenase 1 Homo sapiens 32-36 20974203-10 2011 Because actinomycin D inhibited HO-1 induction by Tg, the induction of HO-1 may be regulated at the transcriptional level. Dactinomycin 8-21 heme oxygenase 1 Homo sapiens 71-75 20974203-10 2011 Because actinomycin D inhibited HO-1 induction by Tg, the induction of HO-1 may be regulated at the transcriptional level. Thapsigargin 50-52 heme oxygenase 1 Homo sapiens 32-36 20974203-10 2011 Because actinomycin D inhibited HO-1 induction by Tg, the induction of HO-1 may be regulated at the transcriptional level. Thapsigargin 50-52 heme oxygenase 1 Homo sapiens 71-75 20974203-11 2011 Moreover, the specific eIF2alpha phosphatase inhibitor salubrinal augmented Tg-induced HO-1 expression. salubrinal 55-65 heme oxygenase 1 Homo sapiens 87-91 20974203-11 2011 Moreover, the specific eIF2alpha phosphatase inhibitor salubrinal augmented Tg-induced HO-1 expression. Thapsigargin 76-78 heme oxygenase 1 Homo sapiens 87-91 20974203-13 2011 On the other hand, the reporter assay revealed that Tg stimulated the antioxidant response element (ARE) that is located in regulatory regions of antioxidant genes such as HO-1. Thapsigargin 52-54 heme oxygenase 1 Homo sapiens 172-176 21765894-2 2011 Heme-oxygenase 1 (HO-1) is known to be the heme inducible isoform, whereas heme-oxygenase 2 (HO-2) is the constitutive enzyme. Heme 43-47 heme oxygenase 1 Homo sapiens 0-16 21765894-2 2011 Heme-oxygenase 1 (HO-1) is known to be the heme inducible isoform, whereas heme-oxygenase 2 (HO-2) is the constitutive enzyme. Heme 43-47 heme oxygenase 1 Homo sapiens 18-22 22203790-0 2011 Quercetin protects primary human osteoblasts exposed to cigarette smoke through activation of the antioxidative enzymes HO-1 and SOD-1. Quercetin 0-9 heme oxygenase 1 Homo sapiens 120-124 22203790-6 2011 Quercetin increased the expression of the anti-oxidative enzymes heme-oxygenase- (HO-) 1 and superoxide-dismutase- (SOD-) 1. Quercetin 0-9 heme oxygenase 1 Homo sapiens 65-88 22203790-7 2011 Inhibiting HO-1 activity abolished the protective effect of Quercetin. Quercetin 60-69 heme oxygenase 1 Homo sapiens 11-15 22203790-9 2011 Quercetin can diminish this damage by scavenging the radicals and by upregulating the expression of HO-1 and SOD-1. Quercetin 0-9 heme oxygenase 1 Homo sapiens 100-104 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Arsenic Trioxide 0-3 heme oxygenase 1 Homo sapiens 32-48 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Arsenic Trioxide 0-3 heme oxygenase 1 Homo sapiens 50-54 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Iron 111-115 heme oxygenase 1 Homo sapiens 32-48 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Iron 111-115 heme oxygenase 1 Homo sapiens 50-54 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. epigallocatechin gallate 120-124 heme oxygenase 1 Homo sapiens 32-48 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. epigallocatechin gallate 120-124 heme oxygenase 1 Homo sapiens 50-54 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Hydrogen Peroxide 133-150 heme oxygenase 1 Homo sapiens 32-48 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Hydrogen Peroxide 133-150 heme oxygenase 1 Homo sapiens 50-54 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Reactive Oxygen Species 215-238 heme oxygenase 1 Homo sapiens 32-48 20950636-7 2011 ATO, on the other hand, induced heme oxygenase-1 (HO-1) to catalyze heme degradation, thereby provided ferrous iron for EGCG-induced hydrogen peroxide to precede Fenton reaction, which in turn generated deleterious reactive oxygen species to damage cell. Reactive Oxygen Species 215-238 heme oxygenase 1 Homo sapiens 50-54 20950636-9 2011 This investigation also provided evidence that ATO, since mainly acted to induce HO-1 in simultaneous treatment with EGCG, could be replaced by other HO-1 inducer with much less human toxicity. Arsenic Trioxide 47-50 heme oxygenase 1 Homo sapiens 81-85 20950636-9 2011 This investigation also provided evidence that ATO, since mainly acted to induce HO-1 in simultaneous treatment with EGCG, could be replaced by other HO-1 inducer with much less human toxicity. Arsenic Trioxide 47-50 heme oxygenase 1 Homo sapiens 150-154 21857081-4 2011 We found that phloretin induced the expression of heme oxygenase-1 (HO-1) protein in a concentration- and time-dependent manner. Phloretin 14-23 heme oxygenase 1 Homo sapiens 50-66 21857081-4 2011 We found that phloretin induced the expression of heme oxygenase-1 (HO-1) protein in a concentration- and time-dependent manner. Phloretin 14-23 heme oxygenase 1 Homo sapiens 68-72 21857081-5 2011 Phloretin induced nuclear factor-E2-related factor 2 (Nrf2) nuclear translocation, and dominant-negative Nrf2 attenuated phloretin-induced expression of HO-1. Phloretin 121-130 heme oxygenase 1 Homo sapiens 153-157 21857081-6 2011 Phloretin activated the JNK, ERK and p38 mitogen-activated protein kinase (MAPK) pathways, and the JNK pathway played an important role in phloretin-induced HO-1 expression. Phloretin 139-148 heme oxygenase 1 Homo sapiens 157-161 21857081-10 2011 These results demonstrate that the expression of HO-1 induced by phloretin is mediated by both the JNK pathway and Nrf2; the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells. Phloretin 65-74 heme oxygenase 1 Homo sapiens 49-53 21857081-10 2011 These results demonstrate that the expression of HO-1 induced by phloretin is mediated by both the JNK pathway and Nrf2; the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells. Cisplatin 145-154 heme oxygenase 1 Homo sapiens 49-53 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Heme 77-81 heme oxygenase 1 Homo sapiens 0-16 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Heme 77-81 heme oxygenase 1 Homo sapiens 18-22 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 0-16 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 18-22 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Iron 104-108 heme oxygenase 1 Homo sapiens 0-16 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Iron 104-108 heme oxygenase 1 Homo sapiens 18-22 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 0-16 21182221-2 2010 Heme oxygenase-1 (HO-1) is a powerful antioxidant enzyme which degrades free heme into biliverdin, free iron and carbon monoxide. Carbon Monoxide 113-128 heme oxygenase 1 Homo sapiens 18-22 20643109-1 2010 Heme oxygenase (HO)-1 is the inducible isoform of the first and rate-limiting enzyme of heme degradation. Heme 88-92 heme oxygenase 1 Homo sapiens 0-21 20643109-4 2010 Beneficial protective effects of HO-1 in inflammation are not only mediated via enzymatic degradation of proinflammatory free heme, but also via production of the anti-inflammatory compounds bilirubin and carbon monoxide. Heme 126-130 heme oxygenase 1 Homo sapiens 33-37 20643109-4 2010 Beneficial protective effects of HO-1 in inflammation are not only mediated via enzymatic degradation of proinflammatory free heme, but also via production of the anti-inflammatory compounds bilirubin and carbon monoxide. Bilirubin 191-200 heme oxygenase 1 Homo sapiens 33-37 20643109-4 2010 Beneficial protective effects of HO-1 in inflammation are not only mediated via enzymatic degradation of proinflammatory free heme, but also via production of the anti-inflammatory compounds bilirubin and carbon monoxide. Carbon Monoxide 205-220 heme oxygenase 1 Homo sapiens 33-37 20837117-6 2010 Importantly, Hg(2+) increased the expression of heme oxygenase-1 (HO-1), a rate limiting enzyme in heme degradation, which coincided with further decrease in the CYP1A1 catalytic activity levels. Heme 48-52 heme oxygenase 1 Homo sapiens 66-70 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Heme 85-89 heme oxygenase 1 Homo sapiens 0-16 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Heme 85-89 heme oxygenase 1 Homo sapiens 18-22 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Iron 98-102 heme oxygenase 1 Homo sapiens 0-16 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Iron 98-102 heme oxygenase 1 Homo sapiens 18-22 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Carbon Monoxide 104-119 heme oxygenase 1 Homo sapiens 0-16 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Carbon Monoxide 104-119 heme oxygenase 1 Homo sapiens 18-22 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Biliverdine 125-135 heme oxygenase 1 Homo sapiens 0-16 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Biliverdine 125-135 heme oxygenase 1 Homo sapiens 18-22 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Bilirubin 182-191 heme oxygenase 1 Homo sapiens 0-16 20713168-4 2010 Heme oxygenase-1 (HO-1), an inducible enzyme, catalyzes the oxidative degradation of heme to free iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Bilirubin 182-191 heme oxygenase 1 Homo sapiens 18-22 20837117-7 2010 Upon using a competitive HO-1 inhibitor, tin mesoporphyrin, heme precursor, hemin, or transfecting the HepG2 cells with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. Polychlorinated Dibenzodioxins 188-192 heme oxygenase 1 Homo sapiens 25-29 20837117-7 2010 Upon using a competitive HO-1 inhibitor, tin mesoporphyrin, heme precursor, hemin, or transfecting the HepG2 cells with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. Polychlorinated Dibenzodioxins 188-192 heme oxygenase 1 Homo sapiens 130-134 20724175-6 2010 DHP-lysine photosensitization induced intracellular oxidative stress, p38 MAPkinase activation, and upregulation of heme oxygenase-1 expression. dhp-lysine 0-10 heme oxygenase 1 Homo sapiens 116-132 20833156-0 2010 Heme oxygenase-1 signals are involved in preferential inhibition of pro-inflammatory cytokine release by surfactin in cells activated with Porphyromonas gingivalis lipopolysaccharide. surfactin peptide 105-114 heme oxygenase 1 Homo sapiens 0-16 20833156-7 2010 We also examined whether the regulatory effect of surfactin on P. gingivalis LPS-stimulated human THP-1 macrophages was mediated by the induction of heme oxygenase-1 (HO-1) signals, and determined that surfactin also induced HO-1 mRNA and protein expression via activation of Nrf-2. surfactin peptide 50-59 heme oxygenase 1 Homo sapiens 149-165 20833156-7 2010 We also examined whether the regulatory effect of surfactin on P. gingivalis LPS-stimulated human THP-1 macrophages was mediated by the induction of heme oxygenase-1 (HO-1) signals, and determined that surfactin also induced HO-1 mRNA and protein expression via activation of Nrf-2. surfactin peptide 50-59 heme oxygenase 1 Homo sapiens 167-171 20833156-8 2010 Additionally, we found that small interfering RNA-mediated knock-down of Nrf-2 significantly inhibited surfactin-induced HO-1 expression. surfactin peptide 103-112 heme oxygenase 1 Homo sapiens 121-125 20833156-9 2010 Furthermore, inhibition of phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK) significantly decreased surfactin-induced HO-1 expression, which is consistent with the suggestion that surfactin-induced HO-1 expression occurs via PI3K/Akt, ERK, and Nrf-2. surfactin peptide 136-145 heme oxygenase 1 Homo sapiens 154-158 20833156-9 2010 Furthermore, inhibition of phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK) significantly decreased surfactin-induced HO-1 expression, which is consistent with the suggestion that surfactin-induced HO-1 expression occurs via PI3K/Akt, ERK, and Nrf-2. surfactin peptide 136-145 heme oxygenase 1 Homo sapiens 234-238 20833156-9 2010 Furthermore, inhibition of phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK) significantly decreased surfactin-induced HO-1 expression, which is consistent with the suggestion that surfactin-induced HO-1 expression occurs via PI3K/Akt, ERK, and Nrf-2. surfactin peptide 216-225 heme oxygenase 1 Homo sapiens 154-158 20833156-9 2010 Furthermore, inhibition of phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK) significantly decreased surfactin-induced HO-1 expression, which is consistent with the suggestion that surfactin-induced HO-1 expression occurs via PI3K/Akt, ERK, and Nrf-2. surfactin peptide 216-225 heme oxygenase 1 Homo sapiens 234-238 20833156-10 2010 Treatment with a selective inhibitor of HO-1 reversed the surfactin-mediated inhibition of pro-inflammatory cytokines, suggesting that surfactin induces anti-inflammatory effects by activating Nrf-2-mediated HO-1 induction via PI3K/Akt and ERK signaling. surfactin peptide 58-67 heme oxygenase 1 Homo sapiens 40-44 20833156-10 2010 Treatment with a selective inhibitor of HO-1 reversed the surfactin-mediated inhibition of pro-inflammatory cytokines, suggesting that surfactin induces anti-inflammatory effects by activating Nrf-2-mediated HO-1 induction via PI3K/Akt and ERK signaling. surfactin peptide 58-67 heme oxygenase 1 Homo sapiens 208-212 20833156-10 2010 Treatment with a selective inhibitor of HO-1 reversed the surfactin-mediated inhibition of pro-inflammatory cytokines, suggesting that surfactin induces anti-inflammatory effects by activating Nrf-2-mediated HO-1 induction via PI3K/Akt and ERK signaling. surfactin peptide 135-144 heme oxygenase 1 Homo sapiens 40-44 20833156-10 2010 Treatment with a selective inhibitor of HO-1 reversed the surfactin-mediated inhibition of pro-inflammatory cytokines, suggesting that surfactin induces anti-inflammatory effects by activating Nrf-2-mediated HO-1 induction via PI3K/Akt and ERK signaling. surfactin peptide 135-144 heme oxygenase 1 Homo sapiens 208-212 21151599-14 2010 FDP-lysine accumulation was associated with the induction of HO-1, no change in GFAP, a decrease in protein levels of the potassium channel subunit Kir4.1, and upregulation of transcripts for VEGF, IL-6, and TNF-alpha. Lysine 4-10 heme oxygenase 1 Homo sapiens 61-65 20446774-0 2010 Nrf2-Mediated heme oxygenase-1 upregulation as adaptive survival response to glucose deprivation-induced apoptosis in HepG2 cells. Glucose 77-84 heme oxygenase 1 Homo sapiens 14-30 20446774-5 2010 To confirm the involvement of Nrf2 in the induction of HO-1 caused by glucose deprivation, we used embryonic fibroblasts prepared from nrf2-(/)-, nrf2(+/)-, and nrf2(+/+) embryos. Glucose 70-77 heme oxygenase 1 Homo sapiens 55-59 20446774-3 2010 Treatment of HepG2 cells with SnCl2, a HO-1 inducer, rescued these cells from glucose deprivation-induced apoptosis, while inhibition of the HO activity with zinc protoporphyrin IX exacerbated apoptosis under the same condition. Glucose 78-85 heme oxygenase 1 Homo sapiens 39-43 20446774-6 2010 Compared to the wild-type and the nrf2(+/)- embryonic fibroblasts, nrf2-(/)- cells were less prone to induce HO-1 expression upon glucose deprivation. Glucose 130-137 heme oxygenase 1 Homo sapiens 109-113 20446774-8 2010 Pretreatment with N-acetylcysteine prevented the glucose deprivation-induced ROS accumulation and also the HO-1 expression. Acetylcysteine 18-34 heme oxygenase 1 Homo sapiens 107-111 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Iron 105-107 heme oxygenase 1 Homo sapiens 459-463 20446774-9 2010 In conclusion, the Nrf2-mediated HO-1 upregulation upon glucose deprivation is mediated by ROS in HepG2 cells, and responsible for the adaptive survival response. Glucose 56-63 heme oxygenase 1 Homo sapiens 33-37 20446774-9 2010 In conclusion, the Nrf2-mediated HO-1 upregulation upon glucose deprivation is mediated by ROS in HepG2 cells, and responsible for the adaptive survival response. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 33-37 20868356-4 2010 HO-1 is the enzyme responsible for the conversion of the heme group to billiverdin, carbon monoxide and iron; a highly regulated cytoprotective enzyme able to respond to numerous chemical or physical stressors, many of which decrease oxygen availability and generate oxidative stress. Heme 57-61 heme oxygenase 1 Homo sapiens 0-4 20868356-4 2010 HO-1 is the enzyme responsible for the conversion of the heme group to billiverdin, carbon monoxide and iron; a highly regulated cytoprotective enzyme able to respond to numerous chemical or physical stressors, many of which decrease oxygen availability and generate oxidative stress. billiverdin 71-82 heme oxygenase 1 Homo sapiens 0-4 20868356-4 2010 HO-1 is the enzyme responsible for the conversion of the heme group to billiverdin, carbon monoxide and iron; a highly regulated cytoprotective enzyme able to respond to numerous chemical or physical stressors, many of which decrease oxygen availability and generate oxidative stress. Carbon Monoxide 84-99 heme oxygenase 1 Homo sapiens 0-4 20868356-4 2010 HO-1 is the enzyme responsible for the conversion of the heme group to billiverdin, carbon monoxide and iron; a highly regulated cytoprotective enzyme able to respond to numerous chemical or physical stressors, many of which decrease oxygen availability and generate oxidative stress. Iron 104-108 heme oxygenase 1 Homo sapiens 0-4 20868356-4 2010 HO-1 is the enzyme responsible for the conversion of the heme group to billiverdin, carbon monoxide and iron; a highly regulated cytoprotective enzyme able to respond to numerous chemical or physical stressors, many of which decrease oxygen availability and generate oxidative stress. Oxygen 234-240 heme oxygenase 1 Homo sapiens 0-4 20704544-2 2010 BVR is the sole catalyst for the conversion of biliverdin-IXalpha the activity product of the stress-inducible HO-1 and the constitutive HO-2, to bilirubin-IXalpha. Biliverdine 47-57 heme oxygenase 1 Homo sapiens 111-115 20704544-2 2010 BVR is the sole catalyst for the conversion of biliverdin-IXalpha the activity product of the stress-inducible HO-1 and the constitutive HO-2, to bilirubin-IXalpha. ixalpha 58-65 heme oxygenase 1 Homo sapiens 111-115 20704544-2 2010 BVR is the sole catalyst for the conversion of biliverdin-IXalpha the activity product of the stress-inducible HO-1 and the constitutive HO-2, to bilirubin-IXalpha. Bilirubin 146-155 heme oxygenase 1 Homo sapiens 111-115 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Heme 33-37 heme oxygenase 1 Homo sapiens 0-16 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 41-56 heme oxygenase 1 Homo sapiens 0-16 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 41-56 heme oxygenase 1 Homo sapiens 18-22 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 58-60 heme oxygenase 1 Homo sapiens 0-16 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 58-60 heme oxygenase 1 Homo sapiens 18-22 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Biliverdine 63-73 heme oxygenase 1 Homo sapiens 0-16 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Biliverdine 63-73 heme oxygenase 1 Homo sapiens 18-22 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Iron 79-91 heme oxygenase 1 Homo sapiens 0-16 20704546-1 2010 Heme oxygenase-1 (HO-1) degrades heme to carbon monoxide (CO), biliverdin, and ferrous iron. Iron 79-91 heme oxygenase 1 Homo sapiens 18-22 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Heme 48-52 heme oxygenase 1 Homo sapiens 0-4 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Iron 56-60 heme oxygenase 1 Homo sapiens 0-4 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Carbon Monoxide 62-77 heme oxygenase 1 Homo sapiens 0-4 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Carbon Monoxide 79-81 heme oxygenase 1 Homo sapiens 0-4 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 0-4 20704548-3 2010 HO-1 catalyzes the degradation of free cellular heme to iron, carbon monoxide (CO) and biliverdin which is eventually converted to bilirubin by biliverdin reductase. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 0-4 20704548-4 2010 In addition to the degradation of free heme, a pro-oxidant, HO-1 exerts anti-oxidant, anti-inflammatory and anti-apoptotic properties via its reaction products. Heme 39-43 heme oxygenase 1 Homo sapiens 60-64 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Heme 45-49 heme oxygenase 1 Homo sapiens 0-16 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Heme 45-49 heme oxygenase 1 Homo sapiens 18-22 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 0-16 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 18-22 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Iron 75-79 heme oxygenase 1 Homo sapiens 0-16 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Iron 75-79 heme oxygenase 1 Homo sapiens 18-22 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 0-16 20704549-1 2010 Heme oxygenase-1 (HO-1), an enzyme degrading heme to carbon monoxide, free iron, and biliverdin, participates in the cell defence against oxidative stress and it has been speculated that it might be a new therapeutic target for neuroprotection. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 18-22 20704549-6 2010 From the therapeutic side, the blood brain barrier represents an obstacle to directly modulate heme oxygenase activity, but drugs activating the transcription actor Nrf2, which have a very diverse molecular structure, may be good candidates to induce HO-1 in concert with other antioxidant and detoxification enzymes. Heme 95-99 heme oxygenase 1 Homo sapiens 251-255 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Heme 36-40 heme oxygenase 1 Homo sapiens 0-16 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Heme 36-40 heme oxygenase 1 Homo sapiens 18-22 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 0-16 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 18-22 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 70-72 heme oxygenase 1 Homo sapiens 0-16 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Carbon Monoxide 70-72 heme oxygenase 1 Homo sapiens 18-22 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Biliverdine 75-85 heme oxygenase 1 Homo sapiens 0-16 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Biliverdine 75-85 heme oxygenase 1 Homo sapiens 18-22 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Iron 91-95 heme oxygenase 1 Homo sapiens 0-16 20875851-8 2010 Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. PNU-282987 13-22 heme oxygenase 1 Homo sapiens 51-67 20875851-8 2010 Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. PNU-282987 13-22 heme oxygenase 1 Homo sapiens 69-73 20875851-8 2010 Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 173-181 heme oxygenase 1 Homo sapiens 51-67 20875851-8 2010 Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 173-181 heme oxygenase 1 Homo sapiens 69-73 20875851-9 2010 The HO-1 inhibitor Sn(IV) protoporphyrin-IX also inhibited the PST/PNU protecting effects. protoporphyrin IX 26-43 heme oxygenase 1 Homo sapiens 4-8 21383490-3 2010 Heme-oxygenase-1 (HO-1) is a rate-limiting enzyme for heme breakdown. Heme 54-58 heme oxygenase 1 Homo sapiens 0-16 21383490-3 2010 Heme-oxygenase-1 (HO-1) is a rate-limiting enzyme for heme breakdown. Heme 54-58 heme oxygenase 1 Homo sapiens 18-22 21383490-4 2010 HO-1 breaks down heme to yield CO, iron and biliverdin. Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 21383490-4 2010 HO-1 breaks down heme to yield CO, iron and biliverdin. Carbon Monoxide 31-33 heme oxygenase 1 Homo sapiens 0-4 21383490-4 2010 HO-1 breaks down heme to yield CO, iron and biliverdin. Iron 35-39 heme oxygenase 1 Homo sapiens 0-4 21383490-4 2010 HO-1 breaks down heme to yield CO, iron and biliverdin. Biliverdine 44-54 heme oxygenase 1 Homo sapiens 0-4 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Iron 99-103 heme oxygenase 1 Homo sapiens 441-457 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Iron 99-103 heme oxygenase 1 Homo sapiens 459-463 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Iron 105-107 heme oxygenase 1 Homo sapiens 441-457 20718734-2 2010 In this study, we have investigated the role of haem arginate (HA) in human male subjects in the modulation of HO-1 expression and its correlation with the GT length polymorphism (GT(n)) in the promoter of the HO-1 gene. heme arginate 48-61 heme oxygenase 1 Homo sapiens 111-115 20718734-2 2010 In this study, we have investigated the role of haem arginate (HA) in human male subjects in the modulation of HO-1 expression and its correlation with the GT length polymorphism (GT(n)) in the promoter of the HO-1 gene. heme arginate 48-61 heme oxygenase 1 Homo sapiens 210-214 20704550-1 2010 Heme oxygenase-1 (HO-1) metabolizes heme to generate carbon monoxide (CO), biliverdin, and iron. Iron 91-95 heme oxygenase 1 Homo sapiens 18-22 20704550-5 2010 The vasoprotection afforded by HO-1 is largely attributable to its end products: CO and the bile pigments, biliverdin and bilirubin. Carbon Monoxide 81-83 heme oxygenase 1 Homo sapiens 31-35 20704550-5 2010 The vasoprotection afforded by HO-1 is largely attributable to its end products: CO and the bile pigments, biliverdin and bilirubin. Biliverdine 107-117 heme oxygenase 1 Homo sapiens 31-35 20704550-5 2010 The vasoprotection afforded by HO-1 is largely attributable to its end products: CO and the bile pigments, biliverdin and bilirubin. Bilirubin 122-131 heme oxygenase 1 Homo sapiens 31-35 20704550-9 2010 Pharmacological induction of HO-1 by heme derivatives, dietary antioxidants, or currently available drugs, is a promising near-term approach, while HO-1 gene delivery is a long-term therapeutic goal. Heme 37-41 heme oxygenase 1 Homo sapiens 29-33 20704552-4 2010 HO-1 degrades heme to biliverdin-IXalpha, carbon monoxide (CO), and iron. Heme 14-18 heme oxygenase 1 Homo sapiens 0-4 20704552-4 2010 HO-1 degrades heme to biliverdin-IXalpha, carbon monoxide (CO), and iron. Biliverdine 22-40 heme oxygenase 1 Homo sapiens 0-4 20704552-4 2010 HO-1 degrades heme to biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 42-57 heme oxygenase 1 Homo sapiens 0-4 20704552-4 2010 HO-1 degrades heme to biliverdin-IXalpha, carbon monoxide (CO), and iron. Carbon Monoxide 59-61 heme oxygenase 1 Homo sapiens 0-4 20704552-4 2010 HO-1 degrades heme to biliverdin-IXalpha, carbon monoxide (CO), and iron. Iron 68-72 heme oxygenase 1 Homo sapiens 0-4 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Copper 113-119 heme oxygenase 1 Homo sapiens 441-457 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Copper 113-119 heme oxygenase 1 Homo sapiens 459-463 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Copper 121-123 heme oxygenase 1 Homo sapiens 441-457 20730621-8 2010 The increased level of oxidative stress in AD brain is reflected by the increased brain content of iron (Fe) and copper (Cu) both capable of stimulating free radical formation (e.g. hydroxyl radicals via Fenton reaction), increased protein and DNA oxidation in the AD brain, enhanced lipid peroxidation, decreased level of cytochrome c oxidase and advanced glycation end products (AGEs), carbonyls, malondialdehyde (MDA), peroxynitrite, and heme oxygenase-1 (HO-1). Copper 121-123 heme oxygenase 1 Homo sapiens 459-463 20851746-12 2010 These findings suggest that CPDT can protect CMNs from THA-induced motor neuron death by activating the Nrf2 pathway and increasing HO-1 protein expression. tha 55-58 heme oxygenase 1 Homo sapiens 132-136 20938215-5 2010 Low PTL concentrations (5 to 10 microM) led to Nrf2-dependent HO-1 induction, which attenuated the apoptogenic effect of PTL in Choi-CK and SCK cells. parthenolide 4-7 heme oxygenase 1 Homo sapiens 62-66 20938215-5 2010 Low PTL concentrations (5 to 10 microM) led to Nrf2-dependent HO-1 induction, which attenuated the apoptogenic effect of PTL in Choi-CK and SCK cells. parthenolide 121-124 heme oxygenase 1 Homo sapiens 62-66 20938215-6 2010 PTL-mediated apoptosis was enhanced by the protein kinase C-alpha inhibitor Ro317549 (Ro) through inhibition of expression and nuclear translocation of Nrf2, resulting in blockage of HO-1 expression. ro317549 76-84 heme oxygenase 1 Homo sapiens 183-187 20938215-6 2010 PTL-mediated apoptosis was enhanced by the protein kinase C-alpha inhibitor Ro317549 (Ro) through inhibition of expression and nuclear translocation of Nrf2, resulting in blockage of HO-1 expression. ro 76-78 heme oxygenase 1 Homo sapiens 183-187 20828541-10 2010 We also demonstrated that the specific ERK inhibitor, PD98059, concentration-dependently blocked on ATX-induced HO-1 expression, and meanwhile PD98059 reversed the protective effect of ATX against Abeta25-35-induced cell death. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 54-61 heme oxygenase 1 Homo sapiens 112-116 20833713-3 2010 We showed that Kupffer cells from ethanol-fed rats and ethanol-treated rat Kupffer cells and THP-1 cells displayed increased mRNA expression of HO-1, NQO1, and hypoxia-inducible factor-1alpha (HIF-1alpha). Ethanol 55-62 heme oxygenase 1 Homo sapiens 144-148 20828541-0 2010 Astaxanthin upregulates heme oxygenase-1 expression through ERK1/2 pathway and its protective effect against beta-amyloid-induced cytotoxicity in SH-SY5Y cells. astaxanthine 0-11 heme oxygenase 1 Homo sapiens 24-40 20828541-8 2010 Moreover, we for the first time have revealed the ATX increased antioxidant enzyme heme oxygenase-1 (HO-1) expression in concentration-dependent and time-dependent manners, which were correlated with its protective effect against Abeta(25-35)-induced injury. astaxanthine 50-53 heme oxygenase 1 Homo sapiens 83-99 20828541-8 2010 Moreover, we for the first time have revealed the ATX increased antioxidant enzyme heme oxygenase-1 (HO-1) expression in concentration-dependent and time-dependent manners, which were correlated with its protective effect against Abeta(25-35)-induced injury. astaxanthine 50-53 heme oxygenase 1 Homo sapiens 101-105 20828541-9 2010 Because the inhibitor of HO-1 activity, ZnPP reversed the protective effect of ATX against Abeta(25-35)-induced cell death. astaxanthine 79-82 heme oxygenase 1 Homo sapiens 25-29 20833713-3 2010 We showed that Kupffer cells from ethanol-fed rats and ethanol-treated rat Kupffer cells and THP-1 cells displayed increased mRNA expression of HO-1, NQO1, and hypoxia-inducible factor-1alpha (HIF-1alpha). Ethanol 34-41 heme oxygenase 1 Homo sapiens 144-148 20828541-10 2010 We also demonstrated that the specific ERK inhibitor, PD98059, concentration-dependently blocked on ATX-induced HO-1 expression, and meanwhile PD98059 reversed the protective effect of ATX against Abeta25-35-induced cell death. astaxanthine 100-103 heme oxygenase 1 Homo sapiens 112-116 20828541-11 2010 Taken together, these findings suggest that astaxanthin can induce HO-1 expression through activation of ERK signal pathways, thereby protecting the SH-SY5Y cells from Abeta(25-35)-induced oxidative cell death. astaxanthine 44-55 heme oxygenase 1 Homo sapiens 67-71 20920477-0 2010 Low concentration of 4-hydroxy hexenal increases heme oxygenase-1 expression through activation of Nrf2 and antioxidative activity in vascular endothelial cells. 4-hydroxyhexenal 21-38 heme oxygenase 1 Homo sapiens 49-65 20679134-4 2010 The goal of this study was to determine whether HO-1 activity could be monitored directly by following BV generation or iron release (using the ferrous iron chelator, ferrozine) in the absence of BVR. ferrous iron chelator 144-165 heme oxygenase 1 Homo sapiens 48-52 21116791-0 2010 Sulforaphane suppresses TARC/CCL17 and MDC/CCL22 expression through heme oxygenase-1 and NF-kappaB in human keratinocytes. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 68-84 21116791-1 2010 Sulforaphane (4-methylsulfinylbutyl isothiocyanate, SFN) from broccoli has been used a chemopreventive photochemical as detoxification of xenobiotics and anti-inflammatory, however, there is no studies for Th2 chemokine expression through heme oxygenase-1 and NF-kappaB in keratinocytes. sulforaphane 0-12 heme oxygenase 1 Homo sapiens 239-255 21116791-1 2010 Sulforaphane (4-methylsulfinylbutyl isothiocyanate, SFN) from broccoli has been used a chemopreventive photochemical as detoxification of xenobiotics and anti-inflammatory, however, there is no studies for Th2 chemokine expression through heme oxygenase-1 and NF-kappaB in keratinocytes. sulforaphane 14-50 heme oxygenase 1 Homo sapiens 239-255 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Oxygen 5-11 heme oxygenase 1 Homo sapiens 18-22 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Heme 108-112 heme oxygenase 1 Homo sapiens 0-16 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Heme 108-112 heme oxygenase 1 Homo sapiens 18-22 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Carbon Monoxide 116-118 heme oxygenase 1 Homo sapiens 0-16 20679134-4 2010 The goal of this study was to determine whether HO-1 activity could be monitored directly by following BV generation or iron release (using the ferrous iron chelator, ferrozine) in the absence of BVR. Ferrozine 167-176 heme oxygenase 1 Homo sapiens 48-52 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Carbon Monoxide 116-118 heme oxygenase 1 Homo sapiens 18-22 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Iron 128-132 heme oxygenase 1 Homo sapiens 0-16 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Iron 128-132 heme oxygenase 1 Homo sapiens 18-22 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Biliverdine 138-148 heme oxygenase 1 Homo sapiens 0-16 20615444-5 2010 The mechanisms of salidroside protected neurons from oxidative stress included the induction of antioxidant enzymes, thioredoxin (Trx), heme oxygenase-1 (HO-1), and peroxiredoxin-I (PrxI); the downregulation of pro-apoptotic protein Bax and the upregulation of anti-apoptotic protein Bcl-X(L). rhodioloside 18-29 heme oxygenase 1 Homo sapiens 136-152 20679134-1 2010 Heme oxygenase 1 (HO-1) uses molecular oxygen and electrons from NADPH cytochrome P450 reductase to convert heme to CO, ferrous iron, and biliverdin (BV). Biliverdine 138-148 heme oxygenase 1 Homo sapiens 18-22 20679134-2 2010 Enzymatic studies with the purified 30-kDa form of HO-1 routinely use a coupled assay containing biliverdin reductase (BVR), which converts BV to bilirubin (BR). Biliverdine 119-121 heme oxygenase 1 Homo sapiens 51-55 20679134-2 2010 Enzymatic studies with the purified 30-kDa form of HO-1 routinely use a coupled assay containing biliverdin reductase (BVR), which converts BV to bilirubin (BR). Bilirubin 146-155 heme oxygenase 1 Homo sapiens 51-55 20679134-4 2010 The goal of this study was to determine whether HO-1 activity could be monitored directly by following BV generation or iron release (using the ferrous iron chelator, ferrozine) in the absence of BVR. Iron 120-124 heme oxygenase 1 Homo sapiens 48-52 20815781-5 2010 Notably, DMSO inhibited methionine sulfoxide reductase activity, induced heme oxygenase-1 (ho-1) mRNA and enhanced oxidant-induced cell death, which indicated that DMSO intensified oxidative stress. Dimethyl Sulfoxide 9-13 heme oxygenase 1 Homo sapiens 73-89 20815781-5 2010 Notably, DMSO inhibited methionine sulfoxide reductase activity, induced heme oxygenase-1 (ho-1) mRNA and enhanced oxidant-induced cell death, which indicated that DMSO intensified oxidative stress. Dimethyl Sulfoxide 9-13 heme oxygenase 1 Homo sapiens 91-95 20815781-5 2010 Notably, DMSO inhibited methionine sulfoxide reductase activity, induced heme oxygenase-1 (ho-1) mRNA and enhanced oxidant-induced cell death, which indicated that DMSO intensified oxidative stress. Dimethyl Sulfoxide 164-168 heme oxygenase 1 Homo sapiens 73-89 20815781-5 2010 Notably, DMSO inhibited methionine sulfoxide reductase activity, induced heme oxygenase-1 (ho-1) mRNA and enhanced oxidant-induced cell death, which indicated that DMSO intensified oxidative stress. Dimethyl Sulfoxide 164-168 heme oxygenase 1 Homo sapiens 91-95 20921523-0 2010 Naive human T cells are activated and proliferate in response to the heme oxygenase-1 inhibitor tin mesoporphyrin. tin mesoporphyrin 96-113 heme oxygenase 1 Homo sapiens 69-85 20921523-3 2010 We demonstrate here that the HO-1 inhibitor tin mesoporphyrin (SnMP) induces activation, proliferation, and maturation of naive CD4(+) and CD8(+) T cells via interactions with CD14(+) monocytes in vitro. tin mesoporphyrin 44-61 heme oxygenase 1 Homo sapiens 29-33 20921523-3 2010 We demonstrate here that the HO-1 inhibitor tin mesoporphyrin (SnMP) induces activation, proliferation, and maturation of naive CD4(+) and CD8(+) T cells via interactions with CD14(+) monocytes in vitro. tin mesoporphyrin 63-67 heme oxygenase 1 Homo sapiens 29-33 21211285-15 2010 The mRNA expressions of Nrf2, HO-1 and NQO1 were increased in LPS + LXA(4) group compared with LPS group (P < 0.05). N-(1H-benzimidazol-2-ylmethyl)-2-methoxyacetamide 68-71 heme oxygenase 1 Homo sapiens 30-34 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Heme 55-59 heme oxygenase 1 Homo sapiens 0-16 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Heme 55-59 heme oxygenase 1 Homo sapiens 18-22 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Iron 93-97 heme oxygenase 1 Homo sapiens 0-16 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Iron 93-97 heme oxygenase 1 Homo sapiens 18-22 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Carbon Monoxide 99-114 heme oxygenase 1 Homo sapiens 0-16 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Carbon Monoxide 99-114 heme oxygenase 1 Homo sapiens 18-22 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Biliverdine 119-129 heme oxygenase 1 Homo sapiens 0-16 20819190-1 2010 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism that converts heme to Fe++, carbon monoxide and biliverdin. Biliverdine 119-129 heme oxygenase 1 Homo sapiens 18-22 20708634-0 2010 GT-repeat polymorphism in the heme oxygenase-1 gene promoter is associated with cardiovascular mortality risk in an arsenic-exposed population in northeastern Taiwan. Arsenic 116-123 heme oxygenase 1 Homo sapiens 30-46 20708634-3 2010 The relationship of HO-1 genotype with arsenic-associated cardiovascular disease has not been studied. Arsenic 39-46 heme oxygenase 1 Homo sapiens 20-24 20708634-4 2010 In this study, we evaluated the relationship between the HO-1 GT-repeat polymorphism and cardiovascular mortality in an arsenic-exposed population. Arsenic 120-127 heme oxygenase 1 Homo sapiens 57-61 20708634-12 2010 Shorter (GT)n repeats in the HO-1 gene promoter may confer protective effects against cardiovascular mortality related to arsenic exposure. Arsenic 122-129 heme oxygenase 1 Homo sapiens 29-33 21223723-8 2010 The HO-1 mRNA expression and the generation of intracellular ROS were downregulated at a specific concentration of H2O2 in the ALDH2 gene transfected group. Hydrogen Peroxide 115-119 heme oxygenase 1 Homo sapiens 4-8 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. organoselenium 9-23 heme oxygenase 1 Homo sapiens 230-246 20961405-0 2010 DMF inhibits PDGF-BB induced airway smooth muscle cell proliferation through induction of heme-oxygenase-1. Dimethyl Fumarate 0-3 heme oxygenase 1 Homo sapiens 90-106 20621084-8 2010 SnPP, a specific inhibitor of HO-1, partly blocked sappanchalcone mediated suppression of inflammatory mediator production, in LPS-stimulated HPDL cells. S-Nitroso-N-propionyl-D,L-penicillamine 0-4 heme oxygenase 1 Homo sapiens 30-34 20621084-8 2010 SnPP, a specific inhibitor of HO-1, partly blocked sappanchalcone mediated suppression of inflammatory mediator production, in LPS-stimulated HPDL cells. sappanchalcone 51-65 heme oxygenase 1 Homo sapiens 30-34 20621084-10 2010 The expression of HO-1 protein by sappanchalcone was significantly reduced by pretreatment with JNK inhibitor. sappanchalcone 34-48 heme oxygenase 1 Homo sapiens 18-22 20621084-11 2010 In conclusion, induction of HO-1 is an important cytoprotective mechanism by which sappanchalcone protects HDP cells from H(2)O(2) and in addition it also exhibits anti-inflammatory effects in LPS-stimulated HPDL cells. sappanchalcone 83-97 heme oxygenase 1 Homo sapiens 28-32 20621084-0 2010 Effects of sappanchalcone on the cytoprotection and anti-inflammation via heme oxygenase-1 in human pulp and periodontal ligament cells. sappanchalcone 11-25 heme oxygenase 1 Homo sapiens 74-90 20621084-4 2010 Sappanchalone concentration proportionately increased heme oxygenase (HO)-1 protein expression and enzyme activity in both HDP and HPDL cells. sappanchalone 0-13 heme oxygenase 1 Homo sapiens 54-75 20621084-6 2010 The cytoprotective effect of sappanchalcone was nullified by HO-1 inhibitor, Tin protoporphyrin (SnPP). sappanchalcone 29-43 heme oxygenase 1 Homo sapiens 61-65 20621084-6 2010 The cytoprotective effect of sappanchalcone was nullified by HO-1 inhibitor, Tin protoporphyrin (SnPP). tin protoporphyrin IX 77-95 heme oxygenase 1 Homo sapiens 61-65 20621084-6 2010 The cytoprotective effect of sappanchalcone was nullified by HO-1 inhibitor, Tin protoporphyrin (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 97-101 heme oxygenase 1 Homo sapiens 61-65 21112485-11 2010 24-hour treatment with 500 mumol/L H2O2 induced remarkable up-regulation of HO-1 and PARP-1 in ARPE-19 cells. Hydrogen Peroxide 35-39 heme oxygenase 1 Homo sapiens 76-80 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. organoselenium 9-23 heme oxygenase 1 Homo sapiens 248-252 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. 3-selena-1-dethiacephem 57-80 heme oxygenase 1 Homo sapiens 230-246 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. 3-selena-1-dethiacephem 57-80 heme oxygenase 1 Homo sapiens 248-252 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. 3-selena-1-dethiacephem 88-111 heme oxygenase 1 Homo sapiens 230-246 20805033-5 2010 Among 42 organoselenium compounds tested, two compounds, 3-selena-1-dethiacephem 13 and 3-selena-1-dethiacephem 14 strongly activated the Nrf2/ARE (antioxidant response element) signaling and thus markedly increased expression of heme oxygenase-1 (HO-1), a phase II antioxidant enzyme. 3-selena-1-dethiacephem 88-111 heme oxygenase 1 Homo sapiens 248-252 20678535-7 2010 Western blot showed that hesperidin facilitated ERK/MAPK phosphorylation which appeared to be responsible for nuclear translocation of Nrf2, thereby inducing cytoprotective heme oxygenase-1 (HO-1) expression. Hesperidin 25-35 heme oxygenase 1 Homo sapiens 173-189 20603117-0 2010 Heme oxygenase 1: another possible target to explain the neuroprotective action of resveratrol, a multifaceted nutrient-based molecule. Resveratrol 83-94 heme oxygenase 1 Homo sapiens 0-16 20603117-5 2010 Resveratrol protects against experimental stroke: putative neuroprotective role of heme oxygenase 1. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 83-99 20603117-9 2010 ), the authors raise a hypothesis that the induction of heme oxygenase 1, an endogenous enzyme that provides resistance against oxidative stress-related neuronal damage, contributes, at least in part, to the neuroprotective action of resveratrol. Resveratrol 234-245 heme oxygenase 1 Homo sapiens 56-72 20709802-0 2010 Adiponectin-mediated heme oxygenase-1 induction protects against iron-induced liver injury via a PPARalpha dependent mechanism. Iron 65-69 heme oxygenase 1 Homo sapiens 21-37 20709802-2 2010 Herein, we show that the deleterious effects of iron dextran on liver function and iron deposition were significantly reversed by adiponectin gene therapy, which was accompanied by AMP-activated protein kinase (AMPK) phosphorylation and heme oxygenase (HO)-1 induction. Iron-Dextran Complex 48-60 heme oxygenase 1 Homo sapiens 237-258 20709802-2 2010 Herein, we show that the deleterious effects of iron dextran on liver function and iron deposition were significantly reversed by adiponectin gene therapy, which was accompanied by AMP-activated protein kinase (AMPK) phosphorylation and heme oxygenase (HO)-1 induction. Iron 48-52 heme oxygenase 1 Homo sapiens 237-258 20605906-5 2010 Sodium nitroprusside (SNP, an NO donor) and 3-(5"-hydroxymethyl-2"-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole 44-91 heme oxygenase 1 Homo sapiens 237-244 20605906-5 2010 Sodium nitroprusside (SNP, an NO donor) and 3-(5"-hydroxymethyl-2"-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). Nitroprusside 0-20 heme oxygenase 1 Homo sapiens 237-244 20605906-5 2010 Sodium nitroprusside (SNP, an NO donor) and 3-(5"-hydroxymethyl-2"-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). 3-(5'-hydroxymethyl-2'-furyl)-1-benzylindazole 44-91 heme oxygenase 1 Homo sapiens 255-262 20605906-5 2010 Sodium nitroprusside (SNP, an NO donor) and 3-(5"-hydroxymethyl-2"-furyl)-1-benzyl indazole [YC-1; acts directly on soluble guanylate cyclase (sGC)] led to further relaxation, yet the residual contraction remained 2 to 3 times higher in H(mox)1(-/-) than H(mox)1(+/+) mice (p < 0.001). Nitroprusside 0-20 heme oxygenase 1 Homo sapiens 255-262 20668435-0 2010 Docosahexaenoic acid suppresses neuroinflammatory responses and induces heme oxygenase-1 expression in BV-2 microglia: implications of antidepressant effects for omega-3 fatty acids. Docosahexaenoic Acids 0-20 heme oxygenase 1 Homo sapiens 72-88 20580034-0 2010 Epigallocatechin gallate-mediated protection against tumor necrosis factor-alpha-induced monocyte chemoattractant protein-1 expression is heme oxygenase-1 dependent. epigallocatechin gallate 0-24 heme oxygenase 1 Homo sapiens 138-154 20580034-3 2010 We hypothesize that flavonoids can down-regulate endothelial inflammatory parameters by modulating HO-1-regulated cell signaling. Flavonoids 20-30 heme oxygenase 1 Homo sapiens 99-103 20580034-4 2010 We focused on the role of HO-1 and its major metabolic product, bilirubin, on mechanisms of tumor necrosis factor-alpha-induced endothelial cell activation and protection by the catechin epigallocatechin gallate (EGCG). catechin epigallocatechin gallate 178-211 heme oxygenase 1 Homo sapiens 26-30 20580034-4 2010 We focused on the role of HO-1 and its major metabolic product, bilirubin, on mechanisms of tumor necrosis factor-alpha-induced endothelial cell activation and protection by the catechin epigallocatechin gallate (EGCG). epigallocatechin gallate 213-217 heme oxygenase 1 Homo sapiens 26-30 20580034-6 2010 Moreover, EGCG up-regulated the expression of HO-1 and further induced the secretion of bilirubin. epigallocatechin gallate 10-14 heme oxygenase 1 Homo sapiens 46-50 20580034-7 2010 The observed anti-inflammatory effects of EGCG were mimicked by the HO-1 inducer cobalt protoporphyrin and abolished by HO-1 gene silencing. epigallocatechin gallate 42-46 heme oxygenase 1 Homo sapiens 68-72 20580034-7 2010 The observed anti-inflammatory effects of EGCG were mimicked by the HO-1 inducer cobalt protoporphyrin and abolished by HO-1 gene silencing. epigallocatechin gallate 42-46 heme oxygenase 1 Homo sapiens 120-124 20580034-7 2010 The observed anti-inflammatory effects of EGCG were mimicked by the HO-1 inducer cobalt protoporphyrin and abolished by HO-1 gene silencing. cobaltiprotoporphyrin 81-102 heme oxygenase 1 Homo sapiens 68-72 20580034-8 2010 These data suggest that the protective properties of flavonoids, such as EGCG, against endothelial inflammation may be regulated in part though induction of HO-1 and subsequent activator protein-1 signaling. Flavonoids 53-63 heme oxygenase 1 Homo sapiens 157-196 20580034-8 2010 These data suggest that the protective properties of flavonoids, such as EGCG, against endothelial inflammation may be regulated in part though induction of HO-1 and subsequent activator protein-1 signaling. epigallocatechin gallate 73-77 heme oxygenase 1 Homo sapiens 157-196 20668435-4 2010 The results of this study showed that DHA reduced expressions of tumor necrosis factor-alpha, interleukin-6, nitric oxide synthase, and cyclo-oxygenase-2, induced by interferon-gamma, and induced upregulation of heme oxygenase-1 (HO-1) in BV-2 microglia. Docosahexaenoic Acids 38-41 heme oxygenase 1 Homo sapiens 212-228 20668435-4 2010 The results of this study showed that DHA reduced expressions of tumor necrosis factor-alpha, interleukin-6, nitric oxide synthase, and cyclo-oxygenase-2, induced by interferon-gamma, and induced upregulation of heme oxygenase-1 (HO-1) in BV-2 microglia. Docosahexaenoic Acids 38-41 heme oxygenase 1 Homo sapiens 230-234 20668435-5 2010 The inhibitory effect of DHA on nitric oxide production was abolished by HO-1 inhibitor zinc protoporphyrin IX. Docosahexaenoic Acids 25-28 heme oxygenase 1 Homo sapiens 73-77 20668435-5 2010 The inhibitory effect of DHA on nitric oxide production was abolished by HO-1 inhibitor zinc protoporphyrin IX. Nitric Oxide 32-44 heme oxygenase 1 Homo sapiens 73-77 20668435-5 2010 The inhibitory effect of DHA on nitric oxide production was abolished by HO-1 inhibitor zinc protoporphyrin IX. protoporphyrin IX 93-110 heme oxygenase 1 Homo sapiens 73-77 20668435-6 2010 In addition, DHA caused AKT and ERK activation in a time-dependent manner, and the DHA-induced HO-1 upregulation could be attenuated by PI-3 kinase/AKT and MEK/ERK inhibitors. Docosahexaenoic Acids 83-86 heme oxygenase 1 Homo sapiens 95-99 20668435-7 2010 DHA also increased IKKalpha/beta phosphorylation, IkappaBalpha phosphorylation, and IkappaBalpha degradation, whereas both nuclear factor-kappaB and IkappaB protease inhibitors could inhibit DHA-induced HO-1 expressions. Docosahexaenoic Acids 0-3 heme oxygenase 1 Homo sapiens 203-207 20668435-8 2010 The other major n-3 PUFA, eicosapentaenoic acid, showed similar effects of DHA on inflammation and HO-1 in repeated key experiments. Eicosapentaenoic Acid 26-47 heme oxygenase 1 Homo sapiens 99-103 20615462-7 2010 Inhibition of gamma-GCS or HO-1 prevented the inhibitory effect of FGF9 on MPP(+)-induced H(2)O(2) production and death in mesencephalic dopaminergic and cortical neurons. mangion-purified polysaccharide (Candida albicans) 75-81 heme oxygenase 1 Homo sapiens 27-31 20732302-0 2010 Altered heme catabolism by heme oxygenase-1 caused by mutations in human NADPH cytochrome P450 reductase. Heme 8-12 heme oxygenase 1 Homo sapiens 27-43 20732302-1 2010 Human heme oxygenase-1 (HO-1) carries out heme catabolism supported by electrons supplied from the NADPH through NADPH P450 reductase (POR, CPR). Heme 6-10 heme oxygenase 1 Homo sapiens 24-28 20732302-1 2010 Human heme oxygenase-1 (HO-1) carries out heme catabolism supported by electrons supplied from the NADPH through NADPH P450 reductase (POR, CPR). NADP 99-104 heme oxygenase 1 Homo sapiens 6-22 20732302-1 2010 Human heme oxygenase-1 (HO-1) carries out heme catabolism supported by electrons supplied from the NADPH through NADPH P450 reductase (POR, CPR). NADP 99-104 heme oxygenase 1 Homo sapiens 24-28 20732302-4 2010 We used purified preparations of wild type and mutant human POR and in vitro reconstitution with purified HO-1 to measure heme degradation in a coupled assay using biliverdin reductase. Heme 122-126 heme oxygenase 1 Homo sapiens 106-110 20732302-5 2010 Here we show that mutations in POR found in patients may reduce HO-1 activity, potentially influencing heme catabolism in individuals carrying mutant POR alleles. Heme 103-107 heme oxygenase 1 Homo sapiens 64-68 20732302-8 2010 Loss of HO-1 activity may result in increased oxidative neurotoxicity, anemia, growth retardation and iron deposition. Iron 102-106 heme oxygenase 1 Homo sapiens 8-12 20599928-5 2010 CAPE increased the expression of Nrf2-dependent luciferase and heme oxygenase-1, a target gene of Nrf2, and elevated the nuclear level of Nrf2 protein, indicating that CAPE activated the Nrf2 pathway. caffeic acid phenethyl ester 168-172 heme oxygenase 1 Homo sapiens 63-79 20846452-4 2010 In this study, the association between malaria disease pathogenicity/severity and (GT)n repeat polymorphism in the promoter region of the inducible HO-1 including the effect of cadmium exposure (potent inducer of HO-1 transcription) as well as polymorphism of TNF were investigated. Cadmium 177-184 heme oxygenase 1 Homo sapiens 148-152 20846452-4 2010 In this study, the association between malaria disease pathogenicity/severity and (GT)n repeat polymorphism in the promoter region of the inducible HO-1 including the effect of cadmium exposure (potent inducer of HO-1 transcription) as well as polymorphism of TNF were investigated. Cadmium 177-184 heme oxygenase 1 Homo sapiens 213-217 20599928-5 2010 CAPE increased the expression of Nrf2-dependent luciferase and heme oxygenase-1, a target gene of Nrf2, and elevated the nuclear level of Nrf2 protein, indicating that CAPE activated the Nrf2 pathway. caffeic acid phenethyl ester 0-4 heme oxygenase 1 Homo sapiens 63-79 20615462-7 2010 Inhibition of gamma-GCS or HO-1 prevented the inhibitory effect of FGF9 on MPP(+)-induced H(2)O(2) production and death in mesencephalic dopaminergic and cortical neurons. Hydrogen Peroxide 90-98 heme oxygenase 1 Homo sapiens 27-31 20822529-4 2010 In this study, we tested the hypothesis that upregulation of HO-1 would inhibit production of the free radical (NO) by interleukin (IL)-1beta-activated human astrocytes. Free Radicals 98-110 heme oxygenase 1 Homo sapiens 61-65 20600671-7 2010 However, sulforaphane could upregulate the expression of HO-1 and NAD(P)H/quinone oxidoreductase-1 (NQO-1) in cells transfected with the empty vector and the wild-type TDP-43. sulforaphane 9-21 heme oxygenase 1 Homo sapiens 57-61 20594940-0 2010 Simvastatin-dependent up-regulation of heme oxygenase-1 via mRNA stabilization in human endothelial cells. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 39-55 20594940-1 2010 Heme oxygenase (HO)-1, which is the inducible isoform of the rate-limiting enzyme of heme degradation, has potent antioxidant and anti-inflammatory effects and is an emerging therapeutic target for the treatment of cardiovascular disease. Heme 85-89 heme oxygenase 1 Homo sapiens 0-21 20594940-3 2010 To further investigate the statin-specific HO-1 regulation, we examined HO-1 gene expression by simvastatin in cell cultures of human endothelial cells. Simvastatin 96-107 heme oxygenase 1 Homo sapiens 72-76 20594940-4 2010 Simvastatin-dependent HO-1 gene activation was significantly reduced by pharmacological inhibition of the p38 MAPK and phosphotidylinositol-3-kinase (PI3K)/Akt pathways. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 22-26 20594940-5 2010 Although HO-1 is considered to be primarily regulated at the transcriptional level, simvastatin induced activity of a human HO-1 promoter gene construct only to a minor extent. Simvastatin 84-95 heme oxygenase 1 Homo sapiens 124-128 20594940-6 2010 By contrast, studies with actinomycin D indicated that the half-life of HO-1 mRNA was significantly prolonged in the presence of simvastatin suggesting a post-transcriptional mode of HO-1 regulation. Dactinomycin 26-39 heme oxygenase 1 Homo sapiens 72-76 20594940-6 2010 By contrast, studies with actinomycin D indicated that the half-life of HO-1 mRNA was significantly prolonged in the presence of simvastatin suggesting a post-transcriptional mode of HO-1 regulation. Dactinomycin 26-39 heme oxygenase 1 Homo sapiens 183-187 20594940-6 2010 By contrast, studies with actinomycin D indicated that the half-life of HO-1 mRNA was significantly prolonged in the presence of simvastatin suggesting a post-transcriptional mode of HO-1 regulation. Simvastatin 129-140 heme oxygenase 1 Homo sapiens 72-76 20594940-6 2010 By contrast, studies with actinomycin D indicated that the half-life of HO-1 mRNA was significantly prolonged in the presence of simvastatin suggesting a post-transcriptional mode of HO-1 regulation. Simvastatin 129-140 heme oxygenase 1 Homo sapiens 183-187 20822529-3 2010 Deleterious free heme is degraded by HO-1 to carbon monoxide, iron and biliverdin, which have potent anti-oxidant and anti-inflammatory properties. Heme 17-21 heme oxygenase 1 Homo sapiens 37-41 20822529-3 2010 Deleterious free heme is degraded by HO-1 to carbon monoxide, iron and biliverdin, which have potent anti-oxidant and anti-inflammatory properties. Carbon Monoxide 45-60 heme oxygenase 1 Homo sapiens 37-41 20822529-3 2010 Deleterious free heme is degraded by HO-1 to carbon monoxide, iron and biliverdin, which have potent anti-oxidant and anti-inflammatory properties. Iron 62-66 heme oxygenase 1 Homo sapiens 37-41 20822529-10 2010 Pretreatment with hemin alone substantially induced both HO-1 mRNA and protein expression, and HO-1 mRNA expression was further enhanced when hemin was combined with IL-1beta treatment. Hemin 18-23 heme oxygenase 1 Homo sapiens 57-61 20822529-3 2010 Deleterious free heme is degraded by HO-1 to carbon monoxide, iron and biliverdin, which have potent anti-oxidant and anti-inflammatory properties. Biliverdine 71-81 heme oxygenase 1 Homo sapiens 37-41 20822529-12 2010 When pretreated with SnPP, the inhibitory effect of hemin on IL-1beta-induced NO production and iNOS expression was reversed, suggesting the involvement of HO-1. S-Nitroso-N-propionyl-D,L-penicillamine 21-25 heme oxygenase 1 Homo sapiens 156-160 20558128-0 2010 Piceatannol induces heme oxygenase-1 expression in human mammary epithelial cells through activation of ARE-driven Nrf2 signaling. 3,3',4,5'-tetrahydroxystilbene 0-11 heme oxygenase 1 Homo sapiens 20-36 20558128-12 2010 The thiol reducing agents, dithiothreitol (100 microM) or beta-mercaptoethanol (1.4 microM), attenuated piceatannol-induced Nrf2 activation and HO-1 expression. Sulfhydryl Compounds 4-9 heme oxygenase 1 Homo sapiens 144-148 20478284-0 2010 High selenium status in individuals exposed to arsenic through coal-burning in Shaanxi (PR of China) modulates antioxidant enzymes, heme oxygenase-1 and DNA damage. Selenium 5-13 heme oxygenase 1 Homo sapiens 132-148 20478284-0 2010 High selenium status in individuals exposed to arsenic through coal-burning in Shaanxi (PR of China) modulates antioxidant enzymes, heme oxygenase-1 and DNA damage. Arsenic 47-54 heme oxygenase 1 Homo sapiens 132-148 20478284-4 2010 RESULTS: High selenium status was correlated with elevated activities of serum superoxide dismutase, glutathione peroxidase, catalase, and reduced levels of malondialdehyde, and increased RNA and protein expression of heme oxygenase-1 in peripheral blood mononuclear cells (PBMC) of individuals in the high arsenic group. Selenium 14-22 heme oxygenase 1 Homo sapiens 218-234 20478284-4 2010 RESULTS: High selenium status was correlated with elevated activities of serum superoxide dismutase, glutathione peroxidase, catalase, and reduced levels of malondialdehyde, and increased RNA and protein expression of heme oxygenase-1 in peripheral blood mononuclear cells (PBMC) of individuals in the high arsenic group. Arsenic 307-314 heme oxygenase 1 Homo sapiens 218-234 20558128-12 2010 The thiol reducing agents, dithiothreitol (100 microM) or beta-mercaptoethanol (1.4 microM), attenuated piceatannol-induced Nrf2 activation and HO-1 expression. Dithiothreitol 27-41 heme oxygenase 1 Homo sapiens 144-148 20558128-12 2010 The thiol reducing agents, dithiothreitol (100 microM) or beta-mercaptoethanol (1.4 microM), attenuated piceatannol-induced Nrf2 activation and HO-1 expression. Mercaptoethanol 58-78 heme oxygenase 1 Homo sapiens 144-148 20558128-12 2010 The thiol reducing agents, dithiothreitol (100 microM) or beta-mercaptoethanol (1.4 microM), attenuated piceatannol-induced Nrf2 activation and HO-1 expression. 3,3',4,5'-tetrahydroxystilbene 104-115 heme oxygenase 1 Homo sapiens 144-148 20558128-13 2010 It is hence likely that piceatannol modifies specific cysteine residues of Keap1, which allows Nrf2 to translocate into the nucleus and bind to ARE, leading to enhancement of the expression of HO-1. 3,3',4,5'-tetrahydroxystilbene 24-35 heme oxygenase 1 Homo sapiens 193-197 20558128-13 2010 It is hence likely that piceatannol modifies specific cysteine residues of Keap1, which allows Nrf2 to translocate into the nucleus and bind to ARE, leading to enhancement of the expression of HO-1. Cysteine 54-62 heme oxygenase 1 Homo sapiens 193-197 20558128-14 2010 The characteristic catechol moiety of piceatannol appears to be critical for induction of Nrf2 activation and subsequent upregulation of HO-1. catechol 19-27 heme oxygenase 1 Homo sapiens 137-141 20558128-14 2010 The characteristic catechol moiety of piceatannol appears to be critical for induction of Nrf2 activation and subsequent upregulation of HO-1. 3,3',4,5'-tetrahydroxystilbene 38-49 heme oxygenase 1 Homo sapiens 137-141 20558128-3 2010 In the present study, we found that piceatannol treatment (30 microM) significantly upregulated the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) and its mRNA transcript at 6h and 3h, respectively in human breast epithelial (MCF10A) cells. 3,3',4,5'-tetrahydroxystilbene 36-47 heme oxygenase 1 Homo sapiens 137-153 20558128-3 2010 In the present study, we found that piceatannol treatment (30 microM) significantly upregulated the expression of the antioxidant enzyme heme oxygenase-1 (HO-1) and its mRNA transcript at 6h and 3h, respectively in human breast epithelial (MCF10A) cells. 3,3',4,5'-tetrahydroxystilbene 36-47 heme oxygenase 1 Homo sapiens 155-159 20558128-6 2010 Upregulation of HO-1 expression by piceatannol through direct binding of Nrf2 to antioxidant response element (ARE) was verified by the chromatin immunoprecipitation (ChIP) assay. 3,3',4,5'-tetrahydroxystilbene 35-46 heme oxygenase 1 Homo sapiens 16-20 20558128-7 2010 siRNA knock down of Nrf2 gene abolished piceatannol-induced HO-1 expression. 3,3',4,5'-tetrahydroxystilbene 40-51 heme oxygenase 1 Homo sapiens 60-64 20558128-8 2010 In addition, piceatannol-induced activation of Nrf2 and/or HO-1 expression was abrogated by the pharmacological inhibitor (LY294002) as well as the kinase-dead form of Akt. 3,3',4,5'-tetrahydroxystilbene 13-24 heme oxygenase 1 Homo sapiens 59-63 20558128-8 2010 In addition, piceatannol-induced activation of Nrf2 and/or HO-1 expression was abrogated by the pharmacological inhibitor (LY294002) as well as the kinase-dead form of Akt. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 123-131 heme oxygenase 1 Homo sapiens 59-63 20558128-9 2010 In an attempt to elucidate the molecular mechanisms underlying cytoprotective or chemoprotective activity exerted by piceatannol, we examined its effect on the signaling pathways responsible for induction of HO-1 expression. 3,3',4,5'-tetrahydroxystilbene 117-128 heme oxygenase 1 Homo sapiens 208-212 20580464-3 2010 A different iron metabolism gene signature was detected in both macrophage types, with the heme regulatory molecules CD163 and Heme Oxygenase-1 (HO-1) being preferentially expressed by M2 (M-CSF) macrophages. Iron 12-16 heme oxygenase 1 Homo sapiens 145-149 20580464-3 2010 A different iron metabolism gene signature was detected in both macrophage types, with the heme regulatory molecules CD163 and Heme Oxygenase-1 (HO-1) being preferentially expressed by M2 (M-CSF) macrophages. Heme 91-95 heme oxygenase 1 Homo sapiens 145-149 20580464-6 2010 In contrast to the HO-1 inhibitor tin protoporphyrin (SnPP), the administration of cobalt protoporphyrin (CoPP), a potent inducer of HO-1 resulted in increased LPS-triggered IL-10 release from M2 (M-CSF) macrophages. cobaltiprotoporphyrin 83-104 heme oxygenase 1 Homo sapiens 133-137 20644008-10 2010 We conclude that estradiol via ER-alpha promotes EPC-mediated capillary formation by a mechanism that involves nongenomic activation of RTKs and HO-1 activation. Estradiol 17-26 heme oxygenase 1 Homo sapiens 145-149 20580464-6 2010 In contrast to the HO-1 inhibitor tin protoporphyrin (SnPP), the administration of cobalt protoporphyrin (CoPP), a potent inducer of HO-1 resulted in increased LPS-triggered IL-10 release from M2 (M-CSF) macrophages. cobaltiprotoporphyrin 106-110 heme oxygenase 1 Homo sapiens 133-137 20644008-2 2010 Because estradiol augments vascular repair, we hypothesize that estradiol increases EPC proliferation and capillary formation via RTK activation and induction of HO-1. Estradiol 8-17 heme oxygenase 1 Homo sapiens 162-166 20644008-2 2010 Because estradiol augments vascular repair, we hypothesize that estradiol increases EPC proliferation and capillary formation via RTK activation and induction of HO-1. Estradiol 64-73 heme oxygenase 1 Homo sapiens 162-166 20644008-7 2010 Estradiol increased HO-1 expression by 2- to 3-fold, an effect blocked by SU5416, and PPT mimicked the effects of estradiol on HO-1. Estradiol 0-9 heme oxygenase 1 Homo sapiens 20-24 20551909-8 2010 At 24 hours postinjury, despite equivalent levels of tubular necrosis, apoptosis, and capillary density between groups, the injection of Ad-HO-1 M phi resulted in preserved renal function (serum creatinine reduced by 46%), and reduced microvascular platelet deposition. Creatinine 195-205 heme oxygenase 1 Homo sapiens 140-144 20644008-7 2010 Estradiol increased HO-1 expression by 2- to 3-fold, an effect blocked by SU5416, and PPT mimicked the effects of estradiol on HO-1. 4,4',4''-(4-propyl-((1)H)-pyrazole-1,3,5-triyl) tris-phenol 86-89 heme oxygenase 1 Homo sapiens 127-131 20644008-7 2010 Estradiol increased HO-1 expression by 2- to 3-fold, an effect blocked by SU5416, and PPT mimicked the effects of estradiol on HO-1. Estradiol 114-123 heme oxygenase 1 Homo sapiens 127-131 20644008-8 2010 The ability of estradiol to enhance capillary formation, increase expression of HO-1, and augment phosphorylation of extracellular signal-regulated kinase 1/2, Akt, and vascular endothelial growth factor receptor 2 was mimicked by its cell-impermeable analog BSA estradiol. Estradiol 15-24 heme oxygenase 1 Homo sapiens 80-84 20502928-2 2010 Two heme oxygenase isoforms, HO-1 and HO-2, exist that differ in several ways, including a complete lack of Cys residues in HO-1 and the presence of three Cys residues as part of heme-regulatory motifs (HRMs) in HO-2. Cysteine 108-111 heme oxygenase 1 Homo sapiens 29-42 20502928-2 2010 Two heme oxygenase isoforms, HO-1 and HO-2, exist that differ in several ways, including a complete lack of Cys residues in HO-1 and the presence of three Cys residues as part of heme-regulatory motifs (HRMs) in HO-2. Cysteine 108-111 heme oxygenase 1 Homo sapiens 29-33 20502928-2 2010 Two heme oxygenase isoforms, HO-1 and HO-2, exist that differ in several ways, including a complete lack of Cys residues in HO-1 and the presence of three Cys residues as part of heme-regulatory motifs (HRMs) in HO-2. Cysteine 155-158 heme oxygenase 1 Homo sapiens 29-42 20502928-2 2010 Two heme oxygenase isoforms, HO-1 and HO-2, exist that differ in several ways, including a complete lack of Cys residues in HO-1 and the presence of three Cys residues as part of heme-regulatory motifs (HRMs) in HO-2. Cysteine 155-158 heme oxygenase 1 Homo sapiens 29-33 20502928-8 2010 These findings are consistent with the presence of a hydrogen-bonding network at the heme"s distal side within the active site of HO-2 with potentially significant differences from that observed in HO-1. Hydrogen 53-61 heme oxygenase 1 Homo sapiens 198-202 20502928-8 2010 These findings are consistent with the presence of a hydrogen-bonding network at the heme"s distal side within the active site of HO-2 with potentially significant differences from that observed in HO-1. Heme 85-89 heme oxygenase 1 Homo sapiens 198-202 20536682-9 2010 The neuroprotective effect was prevented by chelerythrine, LY294002, and Sn (IV) protoporphyrin IX dichloride (SnPP), indicating the participation of the PKC/PI3K/Akt activation and induction of the antioxidant enzyme heme oxygenase-1. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 59-67 heme oxygenase 1 Homo sapiens 218-234 20536682-9 2010 The neuroprotective effect was prevented by chelerythrine, LY294002, and Sn (IV) protoporphyrin IX dichloride (SnPP), indicating the participation of the PKC/PI3K/Akt activation and induction of the antioxidant enzyme heme oxygenase-1. S-Nitroso-N-propionyl-D,L-penicillamine 111-115 heme oxygenase 1 Homo sapiens 218-234 20838567-6 2010 In addition, Western blots showed that ATX elevated of heme oxygenase-1 (HO-1; Hsp32) and Hsp70 protein levels in in vitro. astaxanthine 39-42 heme oxygenase 1 Homo sapiens 55-77 20838567-6 2010 In addition, Western blots showed that ATX elevated of heme oxygenase-1 (HO-1; Hsp32) and Hsp70 protein levels in in vitro. astaxanthine 39-42 heme oxygenase 1 Homo sapiens 79-84 20551909-6 2010 In vitro Ad-HO-1 M phi showed an anti-inflammatory phenotype with increased phagocytosis of apoptotic cells (ACs) and increased interleukin (IL)-10 but reduced TNF-alpha and nitric oxide (NO) following lipopolysaccharide/interferon-gamma (IFN gamma) stimulation compared to control transduced or unmodified M phi. Nitric Oxide 174-186 heme oxygenase 1 Homo sapiens 12-16 20638469-7 2010 An induction of HO-1 expression was observed from concentrations above 5 microM of 4-HNE. 4-hydroxy-2-nonenal 83-88 heme oxygenase 1 Homo sapiens 16-20 20539916-3 2010 One of the proteins important for neovascularisation is heme oxygenase-1 (HO-1), an enzyme degrading heme. Heme 56-60 heme oxygenase 1 Homo sapiens 74-78 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Heme 88-92 heme oxygenase 1 Homo sapiens 12-28 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Heme 88-92 heme oxygenase 1 Homo sapiens 30-34 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 12-28 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 30-34 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Bilirubin 115-124 heme oxygenase 1 Homo sapiens 12-28 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Bilirubin 115-124 heme oxygenase 1 Homo sapiens 30-34 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Iron 135-139 heme oxygenase 1 Homo sapiens 12-28 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Iron 135-139 heme oxygenase 1 Homo sapiens 30-34 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Iron 141-143 heme oxygenase 1 Homo sapiens 12-28 20689484-1 2010 OBJECTIVES: Heme oxygenase 1 (HO-1) is rapidly induced by stress, degrading pro-oxidant heme into carbon monoxide, bilirubin, and free iron (Fe). Iron 141-143 heme oxygenase 1 Homo sapiens 30-34 20689484-4 2010 METHODS: Changes in the HO-1 activity after AMI were analyzed by measuring serum levels of bilirubin and Fe. Bilirubin 91-100 heme oxygenase 1 Homo sapiens 24-28 20689484-9 2010 Furthermore, the serum HO-1 protein level was elevated, and its change was significantly correlated with the change in bilirubin level (r = 0.82, P < 0.005). Bilirubin 119-128 heme oxygenase 1 Homo sapiens 23-27 20796278-0 2010 GT-repeat polymorphism in the heme oxygenase-1 gene promoter and the risk of carotid atherosclerosis related to arsenic exposure. Arsenic 112-119 heme oxygenase 1 Homo sapiens 30-46 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Heme 67-71 heme oxygenase 1 Homo sapiens 12-28 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Heme 67-71 heme oxygenase 1 Homo sapiens 30-34 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Carbon Monoxide 77-92 heme oxygenase 1 Homo sapiens 12-28 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Carbon Monoxide 77-92 heme oxygenase 1 Homo sapiens 30-34 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Biliverdine 94-104 heme oxygenase 1 Homo sapiens 12-28 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Biliverdine 94-104 heme oxygenase 1 Homo sapiens 30-34 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 12-28 20832528-1 2010 BACKGROUND: Heme oxygenase-1 (HO-1) is the enzyme that catabolizes heme into carbon monoxide, biliverdin, and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 30-34 20796278-1 2010 BACKGROUND: Arsenic is a strong stimulus of heme oxygenase (HO)-1 expression in experimental studies in response to oxidative stress caused by a stimulus. Arsenic 12-19 heme oxygenase 1 Homo sapiens 44-65 20796278-5 2010 In this study, we investigated the relationship between HO-1 genetic polymorphism and the risk of atherosclerosis related to arsenic. Arsenic 125-132 heme oxygenase 1 Homo sapiens 56-60 20796278-13 2010 CONCLUSIONS: This exploratory study suggests that at a relatively high level of arsenic exposure, carriers of the short (GT)n allele (< 27 repeats) in the HO-1 gene promoter had a lower probability of developing carotid atherosclerosis than non-carriers of the allele after long-term arsenic exposure via ground water. Arsenic 80-87 heme oxygenase 1 Homo sapiens 158-162 20796278-13 2010 CONCLUSIONS: This exploratory study suggests that at a relatively high level of arsenic exposure, carriers of the short (GT)n allele (< 27 repeats) in the HO-1 gene promoter had a lower probability of developing carotid atherosclerosis than non-carriers of the allele after long-term arsenic exposure via ground water. Arsenic 287-294 heme oxygenase 1 Homo sapiens 158-162 20796278-13 2010 CONCLUSIONS: This exploratory study suggests that at a relatively high level of arsenic exposure, carriers of the short (GT)n allele (< 27 repeats) in the HO-1 gene promoter had a lower probability of developing carotid atherosclerosis than non-carriers of the allele after long-term arsenic exposure via ground water. Water 315-320 heme oxygenase 1 Homo sapiens 158-162 20796278-14 2010 The short (GT)n repeat in the HO-1 gene promoter may provide protective effects against carotid atherosclerosis in individuals with a high level of arsenic exposure. Arsenic 148-155 heme oxygenase 1 Homo sapiens 30-34 20511222-6 2010 Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Hydrogen Peroxide 15-32 heme oxygenase 1 Homo sapiens 110-126 20811623-5 2010 Plasma HO-1 concentrations were significantly correlated with plasma glucose concentrations, HOMA-beta and HOMA-IR (P<0.001). Glucose 69-76 heme oxygenase 1 Homo sapiens 7-11 20511222-6 2010 Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Hydrogen Peroxide 15-32 heme oxygenase 1 Homo sapiens 128-131 20511222-6 2010 Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Hydrogen Peroxide 34-42 heme oxygenase 1 Homo sapiens 110-126 20511222-6 2010 Treatment with hydrogen peroxide (H(2)O(2)) enhances the nuclear colocalization of NRF2 and NRP/B and induces heme oxygenase 1 (HO1). Hydrogen Peroxide 34-42 heme oxygenase 1 Homo sapiens 128-131 20515663-4 2010 Zinc protoporphyrin IX (ZnPP IX), a HO-1 inhibitor, attenuated thrombin-induced injury of cortical cells in a concentration-dependent manner (0.3-3 microM) and tended to inhibit shrinkage of the striatal tissue at 0.3 microM. zinc protoporphyrin 0-22 heme oxygenase 1 Homo sapiens 36-40 20515663-4 2010 Zinc protoporphyrin IX (ZnPP IX), a HO-1 inhibitor, attenuated thrombin-induced injury of cortical cells in a concentration-dependent manner (0.3-3 microM) and tended to inhibit shrinkage of the striatal tissue at 0.3 microM. zinc protoporphyrin 24-31 heme oxygenase 1 Homo sapiens 36-40 20515663-6 2010 The increase of HO-1 protein was suppressed by a p38 MAPK inhibitor SB203580, and early activation of p38 MAPK after thrombin treatment was observed in neurons and microglia in the striatum. SB 203580 68-76 heme oxygenase 1 Homo sapiens 16-20 20515663-10 2010 Thrombin-induced cortical injury may be also regulated by the expression of HO-1 and the resultant production of heme degradation products such as carbon monoxide. Carbon Monoxide 147-162 heme oxygenase 1 Homo sapiens 76-80 20430097-6 2010 Pretreatment with the mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor, U0126, inhibited CaMKII-induced stimulation of HO-1 promoter activity, suggesting that ERK1/2 is a downstream mediator of CaMKII in BDMC signaling to HO-1 expression. U 0126 86-91 heme oxygenase 1 Homo sapiens 133-137 20504881-0 2010 Parenteral iron formulations differentially affect MCP-1, HO-1, and NGAL gene expression and renal responses to injury. Iron 11-15 heme oxygenase 1 Homo sapiens 58-62 20504881-12 2010 We conclude that 1) parenteral iron formulations that stimulate redox signaling can evoke cyto/nephrotoxicity; 2) secondary adaptive responses to this injury (e.g., HO-1/NGAL induction) can initiate a renal tubular cytoresistant state; this suggests a potential new clinical application for intravenous Fe therapy; and 3) FMX is bioneutral regarding these responses. Iron 31-35 heme oxygenase 1 Homo sapiens 165-169 20508205-5 2010 The presence of the S allele led to higher basal HO-1 expression and stronger induction in response to cobalt protoporphyrin, prostaglandin-J(2), hydrogen peroxide, and lipopolysaccharide. Hydrogen Peroxide 146-163 heme oxygenase 1 Homo sapiens 49-53 20806083-0 2010 Resveratrol protects human lens epithelial cells against H2O2-induced oxidative stress by increasing catalase, SOD-1, and HO-1 expression. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 122-126 20806083-0 2010 Resveratrol protects human lens epithelial cells against H2O2-induced oxidative stress by increasing catalase, SOD-1, and HO-1 expression. Hydrogen Peroxide 57-61 heme oxygenase 1 Homo sapiens 122-126 20806083-9 2010 RESULTS: Resveratrol clearly reduced H(2)O(2) induced cell apoptosis and ROS accumulation; protected HLEB-3 cells from H(2)O(2) induced oxidative damage, and increased the expression levels of SOD-1, catalase, and HO-1. Resveratrol 9-20 heme oxygenase 1 Homo sapiens 214-218 20430097-0 2010 Up-regulation of heme oxygenase-1 expression through CaMKII-ERK1/2-Nrf2 signaling mediates the anti-inflammatory effect of bisdemethoxycurcumin in LPS-stimulated macrophages. bisdemethoxycurcumin 123-143 heme oxygenase 1 Homo sapiens 17-33 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. bisdemethoxycurcumin 134-154 heme oxygenase 1 Homo sapiens 95-111 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. bisdemethoxycurcumin 134-154 heme oxygenase 1 Homo sapiens 113-117 20430097-6 2010 Pretreatment with the mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor, U0126, inhibited CaMKII-induced stimulation of HO-1 promoter activity, suggesting that ERK1/2 is a downstream mediator of CaMKII in BDMC signaling to HO-1 expression. U 0126 86-91 heme oxygenase 1 Homo sapiens 236-240 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. bisdemethoxycurcumin 156-160 heme oxygenase 1 Homo sapiens 95-111 20430097-6 2010 Pretreatment with the mitogen-activated protein kinase kinase 1/2 (MEK1/2) inhibitor, U0126, inhibited CaMKII-induced stimulation of HO-1 promoter activity, suggesting that ERK1/2 is a downstream mediator of CaMKII in BDMC signaling to HO-1 expression. bisdemethoxycurcumin 218-222 heme oxygenase 1 Homo sapiens 133-137 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. bisdemethoxycurcumin 156-160 heme oxygenase 1 Homo sapiens 113-117 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. Curcumin 146-154 heme oxygenase 1 Homo sapiens 95-111 20430097-9 2010 Collectively, our findings identify a Ca(2+)/calmodulin-CaMKII-ERK1/2-Nrf2 cascade as a novel anti-inflammatory pathway mediating BDMC signaling to HO-1 expression in macrophages. bisdemethoxycurcumin 130-134 heme oxygenase 1 Homo sapiens 148-152 20430097-1 2010 We have identified a novel anti-inflammatory signaling pathway that leads to the expression of heme oxygenase-1 (HO-1) in response to bisdemethoxycurcumin (BDMC), an analog of curcumin. Curcumin 146-154 heme oxygenase 1 Homo sapiens 113-117 20430097-4 2010 The signaling pathway involved in BDMC-mediated HO-1 induction included Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) and extracellular signal-regulated kinase 1/2 (ERK1/2). bisdemethoxycurcumin 34-38 heme oxygenase 1 Homo sapiens 48-52 20647090-4 2010 RESULTS: HO-1 induction by cobaltic protoporphyrin IX (CoPP) in HDPCs increased cell growth and mineralization and up-regulated the messenger RNA expression of such odontoblastic markers as alkaline phosphatase, osteopontin, bone sialoprotein, dentin matrix protein-1, and dentin sialophosphoprotein. cobaltic protoporphyrin ix 27-53 heme oxygenase 1 Homo sapiens 9-13 20722927-2 2010 We investigated the activation of HO-1 by whisky, which contains various phenolic substances. whisky 42-48 heme oxygenase 1 Homo sapiens 34-38 20647090-4 2010 RESULTS: HO-1 induction by cobaltic protoporphyrin IX (CoPP) in HDPCs increased cell growth and mineralization and up-regulated the messenger RNA expression of such odontoblastic markers as alkaline phosphatase, osteopontin, bone sialoprotein, dentin matrix protein-1, and dentin sialophosphoprotein. copp 55-59 heme oxygenase 1 Homo sapiens 9-13 20647090-6 2010 CONCLUSIONS: CoPP treatment results in dental pulp cell proliferation and odontoblast differentiation that appears partly mediated by HO-1. copp 13-17 heme oxygenase 1 Homo sapiens 134-138 20722927-4 2010 To determine the compounds with potent HO-1-inducing activity among the whisky congeners, several chemicals that had been reported to exist in whisky or oak barrels were screened, and coniferyl aldehyde and sinapyl aldehyde showed the activity. coniferaldehyde 184-202 heme oxygenase 1 Homo sapiens 39-43 20722927-4 2010 To determine the compounds with potent HO-1-inducing activity among the whisky congeners, several chemicals that had been reported to exist in whisky or oak barrels were screened, and coniferyl aldehyde and sinapyl aldehyde showed the activity. sinapyl aldehyde 207-223 heme oxygenase 1 Homo sapiens 39-43 20430037-0 2010 Conversion of biliverdin to bilirubin by biliverdin reductase contributes to endothelial cell protection by heme oxygenase-1-evidence for direct and indirect antioxidant actions of bilirubin. Biliverdine 14-24 heme oxygenase 1 Homo sapiens 108-124 20430037-0 2010 Conversion of biliverdin to bilirubin by biliverdin reductase contributes to endothelial cell protection by heme oxygenase-1-evidence for direct and indirect antioxidant actions of bilirubin. Bilirubin 28-37 heme oxygenase 1 Homo sapiens 108-124 20430037-0 2010 Conversion of biliverdin to bilirubin by biliverdin reductase contributes to endothelial cell protection by heme oxygenase-1-evidence for direct and indirect antioxidant actions of bilirubin. Bilirubin 181-190 heme oxygenase 1 Homo sapiens 108-124 20430037-2 2010 HO-1-derived bilirubin is an efficient scavenger of reactive oxygen and nitrogen species (RONS). Bilirubin 13-22 heme oxygenase 1 Homo sapiens 0-4 20430037-2 2010 HO-1-derived bilirubin is an efficient scavenger of reactive oxygen and nitrogen species (RONS). reactive oxygen and nitrogen species 52-88 heme oxygenase 1 Homo sapiens 0-4 20430037-2 2010 HO-1-derived bilirubin is an efficient scavenger of reactive oxygen and nitrogen species (RONS). rons 90-94 heme oxygenase 1 Homo sapiens 0-4 20430037-3 2010 It remains to determine whether conversion of biliverdin to bilirubin is an essential step for HO-1-conferred protection of endothelial cells. Biliverdine 46-56 heme oxygenase 1 Homo sapiens 95-99 20486808-5 2010 HO-1 was induced by infection with a recombinant adenovirus carrying the human HO-1 gene or an inducer of HO-1 activity, cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 121-145 heme oxygenase 1 Homo sapiens 0-4 20430037-3 2010 It remains to determine whether conversion of biliverdin to bilirubin is an essential step for HO-1-conferred protection of endothelial cells. Bilirubin 60-69 heme oxygenase 1 Homo sapiens 95-99 20430037-7 2010 HO-1- and BVR-silenced cells have increased levels of oxidative stress and bilirubin but not biliverdin increased expression of the protective protein GTP-cyclohydrolase. Bilirubin 75-84 heme oxygenase 1 Homo sapiens 0-4 20430037-8 2010 Moreover, protection by hemin-induced HO-1 expression or biliverdin-triggered bilirubin formation was impaired upon silencing of the HO-1 or BVR gene, respectively. Biliverdine 57-67 heme oxygenase 1 Homo sapiens 133-137 20430037-8 2010 Moreover, protection by hemin-induced HO-1 expression or biliverdin-triggered bilirubin formation was impaired upon silencing of the HO-1 or BVR gene, respectively. Bilirubin 78-87 heme oxygenase 1 Homo sapiens 133-137 20486808-5 2010 HO-1 was induced by infection with a recombinant adenovirus carrying the human HO-1 gene or an inducer of HO-1 activity, cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 147-151 heme oxygenase 1 Homo sapiens 0-4 20471992-9 2010 The HO-1 competitive inhibitor zinc protoporphyrin partially but significantly restored the sensitivity of precultured monocytes to CBD-mediated apoptosis. zinc protoporphyrin 31-50 heme oxygenase 1 Homo sapiens 4-8 20536138-7 2010 In parallel, andrographolide significantly induced the expression of HO-1 in a concentration-dependent fashion (p < 0.05). andrographolide 13-28 heme oxygenase 1 Homo sapiens 69-73 20501657-3 2010 Oxidative stress induced by H(2)O(2) led to an increase in MARE activity and expression of heme oxygenase-1 (HO-1), an inducible antioxidant defense enzyme. Hydrogen Peroxide 28-36 heme oxygenase 1 Homo sapiens 91-107 20501657-3 2010 Oxidative stress induced by H(2)O(2) led to an increase in MARE activity and expression of heme oxygenase-1 (HO-1), an inducible antioxidant defense enzyme. Hydrogen Peroxide 28-36 heme oxygenase 1 Homo sapiens 109-113 20501657-6 2010 In contrast, Bach1 overexpression abolished HO-1 induction by H(2)O(2), which led to increased reactive oxygen species accumulation. Hydrogen Peroxide 62-70 heme oxygenase 1 Homo sapiens 44-48 20501657-6 2010 In contrast, Bach1 overexpression abolished HO-1 induction by H(2)O(2), which led to increased reactive oxygen species accumulation. Reactive Oxygen Species 95-118 heme oxygenase 1 Homo sapiens 44-48 20599745-0 2010 Celastrol suppresses IFN-gamma-induced ICAM-1 expression and subsequent monocyte adhesiveness via the induction of heme oxygenase-1 in the HaCaT cells. celastrol 0-9 heme oxygenase 1 Homo sapiens 115-131 20599745-3 2010 We found that celastrol induced mRNA and protein expression of heme oxygenase-1 (HO-1) in the human keratinocyte cell line HaCaT. celastrol 14-23 heme oxygenase 1 Homo sapiens 63-79 20599745-3 2010 We found that celastrol induced mRNA and protein expression of heme oxygenase-1 (HO-1) in the human keratinocyte cell line HaCaT. celastrol 14-23 heme oxygenase 1 Homo sapiens 81-85 20599745-4 2010 Treatment of HaCaT cells with tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, reversed the suppressive effect of celastrol on IFN-gamma-induced protein and mRNA expression of ICAM-1. tin protoporphyrin IX 30-51 heme oxygenase 1 Homo sapiens 84-88 20599745-4 2010 Treatment of HaCaT cells with tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, reversed the suppressive effect of celastrol on IFN-gamma-induced protein and mRNA expression of ICAM-1. S-Nitroso-N-propionyl-D,L-penicillamine 53-57 heme oxygenase 1 Homo sapiens 84-88 20599745-4 2010 Treatment of HaCaT cells with tin protoporphyrin IX (SnPP), a specific inhibitor of HO-1, reversed the suppressive effect of celastrol on IFN-gamma-induced protein and mRNA expression of ICAM-1. celastrol 125-134 heme oxygenase 1 Homo sapiens 84-88 20599745-5 2010 HO-1 knockdown using small interfering RNA (siRNA) led to reverse inhibition of IFN-gamma-induced up-regulation of ICAM-1 by celastrol. celastrol 125-134 heme oxygenase 1 Homo sapiens 0-4 20599745-7 2010 Furthermore, blockage of HO-1 activity by SnPP and HO-1 siRNA reversed the inhibitory effect of celastrol on IFN-gamma-induced adhesion of monocytes to keratinocytes. S-Nitroso-N-propionyl-D,L-penicillamine 42-46 heme oxygenase 1 Homo sapiens 25-29 20599745-7 2010 Furthermore, blockage of HO-1 activity by SnPP and HO-1 siRNA reversed the inhibitory effect of celastrol on IFN-gamma-induced adhesion of monocytes to keratinocytes. celastrol 96-105 heme oxygenase 1 Homo sapiens 25-29 20599745-7 2010 Furthermore, blockage of HO-1 activity by SnPP and HO-1 siRNA reversed the inhibitory effect of celastrol on IFN-gamma-induced adhesion of monocytes to keratinocytes. celastrol 96-105 heme oxygenase 1 Homo sapiens 51-55 20498371-8 2010 Also, 8-nitro-cGMP caused S-guanylation of KEAP1 in cells, which led to Nrf2 activation and subsequent induction of antioxidant enzymes, including heme oxygenase-1; thus, 8-nitro-cGMP protected cells against cytotoxic effects of hydrogen peroxide. 8-nitroguanosine 3',5'-cyclic monophosphate 6-18 heme oxygenase 1 Homo sapiens 147-163 20498371-8 2010 Also, 8-nitro-cGMP caused S-guanylation of KEAP1 in cells, which led to Nrf2 activation and subsequent induction of antioxidant enzymes, including heme oxygenase-1; thus, 8-nitro-cGMP protected cells against cytotoxic effects of hydrogen peroxide. 8-nitroguanosine 3',5'-cyclic monophosphate 171-183 heme oxygenase 1 Homo sapiens 147-163 20498371-8 2010 Also, 8-nitro-cGMP caused S-guanylation of KEAP1 in cells, which led to Nrf2 activation and subsequent induction of antioxidant enzymes, including heme oxygenase-1; thus, 8-nitro-cGMP protected cells against cytotoxic effects of hydrogen peroxide. Hydrogen Peroxide 229-246 heme oxygenase 1 Homo sapiens 147-163 20676377-2 2010 However, HO-1 protein levels are increased in dopaminergic neurons of Parkinson"s disease (PD) patients, suggesting its participation in free-iron deposition, oxidative stress and neurotoxicity. Iron 142-146 heme oxygenase 1 Homo sapiens 9-13 20599745-8 2010 These results suggest that celastrol may exert anti-inflammatory responses by suppressing IFN-gamma-induced expression of ICAM-1 and subsequent monocyte adhesion via expression of HO-1 in the keratinocytes. celastrol 27-36 heme oxygenase 1 Homo sapiens 180-184 20536138-9 2010 Transfection with HO-1-specific small interfering RNA knocked down HO-1 expression, and the inhibition of expression of ICAM-1 by andrographolide was significantly reversed. andrographolide 130-145 heme oxygenase 1 Homo sapiens 18-22 20536138-10 2010 These results suggest that stimulation of Nrf2-dependent HO-1 expression is involved in the suppression of TNF-alpha-induced ICAM-1 expression exerted by andrographolide. andrographolide 154-169 heme oxygenase 1 Homo sapiens 57-61 20618971-9 2010 Pretreatment of cells with wortmannin inhibited translocation of Nrf2 and induction of HMOX1. Wortmannin 27-37 heme oxygenase 1 Homo sapiens 87-92 20618971-10 2010 Wortmannin pretreatment was also able to diminish adaphostin induction of HMOX1, and as a consequence, enhance the toxicity of adaphostin to NCI-H522. Wortmannin 0-10 heme oxygenase 1 Homo sapiens 74-79 20618971-10 2010 Wortmannin pretreatment was also able to diminish adaphostin induction of HMOX1, and as a consequence, enhance the toxicity of adaphostin to NCI-H522. NSC 680410 50-60 heme oxygenase 1 Homo sapiens 74-79 20618971-11 2010 CONCLUSIONS: Adaphostin-induced oxidative stress in NCI-H522 was mediated through nuclear translocation of Nrf2 leading to upregulation of HMOX1. NSC 680410 13-23 heme oxygenase 1 Homo sapiens 139-144 20593496-1 2010 Heme oxygenase (HO)-1 is the inducible isoform of the first and rate-limiting enzyme of heme degradation. Heme 88-92 heme oxygenase 1 Homo sapiens 0-21 20461740-0 2010 Resveratrol inhibits human lung adenocarcinoma cell metastasis by suppressing heme oxygenase 1-mediated nuclear factor-kappaB pathway and subsequently downregulating expression of matrix metalloproteinases. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 78-94 20378845-5 2010 In the interactions between hemoglobin and atheroma lipids, hemoglobin and heme promote further lipid oxidation and subsequently endothelial reactions such as upregulation of heme oxygenase-1 and cytotoxicity to endothelium. Heme 75-79 heme oxygenase 1 Homo sapiens 175-191 20664735-6 2010 After 24 h exposure of Huh7 cells to GLA, we identified several genes encoding the antioxidant proteins to be upregulated: heme oxygenase-1 (HO-1), aldo-keto reductase 1 family C1 (AKR1C1), C4 (AKR1C4), and thioredoxin (Trx). gamma-Linolenic Acid 37-40 heme oxygenase 1 Homo sapiens 123-139 20664735-6 2010 After 24 h exposure of Huh7 cells to GLA, we identified several genes encoding the antioxidant proteins to be upregulated: heme oxygenase-1 (HO-1), aldo-keto reductase 1 family C1 (AKR1C1), C4 (AKR1C4), and thioredoxin (Trx). gamma-Linolenic Acid 37-40 heme oxygenase 1 Homo sapiens 141-145 20664735-7 2010 The HO-1 protein levels were overexpressed in Huh7 cells after GLA exposure using a Western blot analysis. gamma-Linolenic Acid 63-66 heme oxygenase 1 Homo sapiens 4-8 20664735-9 2010 GLA treatment has induced cell growth inhibition, ROS generation including lipid peroxidation, and HO-1 production for antioxidant protection against oxidative stress caused by GLA in Huh7 cells. gamma-Linolenic Acid 0-3 heme oxygenase 1 Homo sapiens 99-103 20664735-9 2010 GLA treatment has induced cell growth inhibition, ROS generation including lipid peroxidation, and HO-1 production for antioxidant protection against oxidative stress caused by GLA in Huh7 cells. gamma-Linolenic Acid 177-180 heme oxygenase 1 Homo sapiens 99-103 20155807-4 2010 MG-132, a proteasome inhibitor, increased the amount of HO-1 protein mainly in astrocytes of primary mesencephalic cultures. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-6 heme oxygenase 1 Homo sapiens 56-60 20155807-5 2010 Quantitative RT-PCR analysis revealed that lactacystin upregulated HO-1 mRNA expression. lactacystin 43-54 heme oxygenase 1 Homo sapiens 67-71 20155807-6 2010 Proteasome inhibition with MG132 also increased the cytomegalovirus promoter-driven expression of Flag-HO-1 protein and resulted in an accumulation of ubiquitinated Flag-HO-1 in Flag-HO-1-overexpressing PC12 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 27-32 heme oxygenase 1 Homo sapiens 103-107 20155807-6 2010 Proteasome inhibition with MG132 also increased the cytomegalovirus promoter-driven expression of Flag-HO-1 protein and resulted in an accumulation of ubiquitinated Flag-HO-1 in Flag-HO-1-overexpressing PC12 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 27-32 heme oxygenase 1 Homo sapiens 170-174 20155807-6 2010 Proteasome inhibition with MG132 also increased the cytomegalovirus promoter-driven expression of Flag-HO-1 protein and resulted in an accumulation of ubiquitinated Flag-HO-1 in Flag-HO-1-overexpressing PC12 cells. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 27-32 heme oxygenase 1 Homo sapiens 170-174 20155807-7 2010 In addition, a cycloheximide chase assay demonstrated that the degradation of Flag-HO-1 protein was slowed by MG-132. Cycloheximide 15-28 heme oxygenase 1 Homo sapiens 83-87 20155807-7 2010 In addition, a cycloheximide chase assay demonstrated that the degradation of Flag-HO-1 protein was slowed by MG-132. Magnesium 110-112 heme oxygenase 1 Homo sapiens 83-87 20155807-9 2010 Proteasome inhibitors protected dopaminergic neurons from 6-hydroxydopamine (6-OHDA)-induced toxicity and this neuroprotection was abrogated by co-treatment with zinc protoporphyrin IX, a HO-1 inhibitor. Oxidopamine 58-75 heme oxygenase 1 Homo sapiens 188-192 20155807-9 2010 Proteasome inhibitors protected dopaminergic neurons from 6-hydroxydopamine (6-OHDA)-induced toxicity and this neuroprotection was abrogated by co-treatment with zinc protoporphyrin IX, a HO-1 inhibitor. Oxidopamine 77-83 heme oxygenase 1 Homo sapiens 188-192 20155807-9 2010 Proteasome inhibitors protected dopaminergic neurons from 6-hydroxydopamine (6-OHDA)-induced toxicity and this neuroprotection was abrogated by co-treatment with zinc protoporphyrin IX, a HO-1 inhibitor. zinc protoporphyrin 162-184 heme oxygenase 1 Homo sapiens 188-192 20155807-10 2010 Furthermore, 6-OHDA-induced toxicity was blocked by bilirubin and carbon monoxide, products of the HO-1-catalyzed degradation of heme. Oxidopamine 13-19 heme oxygenase 1 Homo sapiens 99-103 20155807-10 2010 Furthermore, 6-OHDA-induced toxicity was blocked by bilirubin and carbon monoxide, products of the HO-1-catalyzed degradation of heme. Bilirubin 52-61 heme oxygenase 1 Homo sapiens 99-103 20155807-10 2010 Furthermore, 6-OHDA-induced toxicity was blocked by bilirubin and carbon monoxide, products of the HO-1-catalyzed degradation of heme. Carbon Monoxide 66-81 heme oxygenase 1 Homo sapiens 99-103 20155807-10 2010 Furthermore, 6-OHDA-induced toxicity was blocked by bilirubin and carbon monoxide, products of the HO-1-catalyzed degradation of heme. Heme 129-133 heme oxygenase 1 Homo sapiens 99-103 20461740-7 2010 Resveratrol significantly inhibited HO-1-mediated MMP-9 (35% inhibition) and MMP-2 (28% inhibition) expression in lung cancer cells. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 36-40 20461740-10 2010 Our results suggest that resveratrol inhibited HO-1 and subsequently MMP-9 and MMP-2 expression in lung cancer cells. Resveratrol 25-36 heme oxygenase 1 Homo sapiens 47-51 20461740-11 2010 The inhibitory effects of resveratrol on MMP expression and invasion of lung cancer cells are, in part, associated with the HO-1-mediated NF-kappaB pathway. Resveratrol 26-37 heme oxygenase 1 Homo sapiens 124-128 20307651-8 2010 In Pin1-VSMCs, heme oxygenase-1 (HO-1) induction in response to nitric oxide donor was suppressed compared to control VSMCs. Nitric Oxide 64-76 heme oxygenase 1 Homo sapiens 15-31 20692427-0 2010 The in vitro protection of human decay accelerating factor and hDAF/heme oxygenase-1 transgenes in porcine aortic endothelial cells against sera of Formosan macaques. formosan 148-156 heme oxygenase 1 Homo sapiens 68-84 20307651-8 2010 In Pin1-VSMCs, heme oxygenase-1 (HO-1) induction in response to nitric oxide donor was suppressed compared to control VSMCs. vsmcs 8-13 heme oxygenase 1 Homo sapiens 15-31 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Heme 43-47 heme oxygenase 1 Homo sapiens 0-16 20303947-1 2010 Heme oxygenase-1 (HO-1), a known cytoprotective enzyme implicated also in the cell cycle regulation and angiogenesis, exerts many of its beneficial effects through carbon monoxide (CO). Carbon Monoxide 164-179 heme oxygenase 1 Homo sapiens 0-16 20303947-1 2010 Heme oxygenase-1 (HO-1), a known cytoprotective enzyme implicated also in the cell cycle regulation and angiogenesis, exerts many of its beneficial effects through carbon monoxide (CO). Carbon Monoxide 164-179 heme oxygenase 1 Homo sapiens 18-22 20226179-0 2010 Structure-activity relationships in the cytoprotective effect of caffeic acid phenethyl ester (CAPE) and fluorinated derivatives: effects on heme oxygenase-1 induction and antioxidant activities. caffeic acid phenethyl ester 65-93 heme oxygenase 1 Homo sapiens 141-157 20226179-0 2010 Structure-activity relationships in the cytoprotective effect of caffeic acid phenethyl ester (CAPE) and fluorinated derivatives: effects on heme oxygenase-1 induction and antioxidant activities. caffeic acid phenethyl ester 95-99 heme oxygenase 1 Homo sapiens 141-157 20226179-0 2010 Structure-activity relationships in the cytoprotective effect of caffeic acid phenethyl ester (CAPE) and fluorinated derivatives: effects on heme oxygenase-1 induction and antioxidant activities. fluorinated 105-116 heme oxygenase 1 Homo sapiens 141-157 20226179-2 2010 To ascertain the involvement of HO-1 induction in the cytoprotective effects of CAPE analogues, their ability to induce HO-1 at 20microM was determined by reverse transcriptase polymerase chain reaction, western blotting and the use of HO-1 inhibitor tin protoporphyrin IX. caffeic acid phenethyl ester 80-84 heme oxygenase 1 Homo sapiens 32-36 20226179-2 2010 To ascertain the involvement of HO-1 induction in the cytoprotective effects of CAPE analogues, their ability to induce HO-1 at 20microM was determined by reverse transcriptase polymerase chain reaction, western blotting and the use of HO-1 inhibitor tin protoporphyrin IX. caffeic acid phenethyl ester 80-84 heme oxygenase 1 Homo sapiens 120-124 20226179-2 2010 To ascertain the involvement of HO-1 induction in the cytoprotective effects of CAPE analogues, their ability to induce HO-1 at 20microM was determined by reverse transcriptase polymerase chain reaction, western blotting and the use of HO-1 inhibitor tin protoporphyrin IX. caffeic acid phenethyl ester 80-84 heme oxygenase 1 Homo sapiens 120-124 20226179-3 2010 There was significant induction of HO-1 by CAPE derivatives. caffeic acid phenethyl ester 43-47 heme oxygenase 1 Homo sapiens 35-39 20226179-6 2010 The maintenance of at least one hydroxyl group on the CAPE catechol ring and the phenethyl ester portion was required for HO-1 induction. Bis(ethylphenyl)ether 81-96 heme oxygenase 1 Homo sapiens 122-126 20226179-10 2010 These results indicate that the induction of HO-1 plays a more important role in the cytoprotective activity of CAPE derivatives than their direct antioxidant activity. caffeic acid phenethyl ester 112-116 heme oxygenase 1 Homo sapiens 45-49 20385560-6 2010 Covalent modification at two conserved cysteine residues, corresponding to Cys(261) and Cys(273) in HDAC1, coincided with attenuation of histone deacetylase activity, changes in histone H3 and H4 acetylation patterns, derepression of a LEF1.beta-catenin model system, and transcription of HDAC-repressed genes, e.g. heme oxygenase-1 (HO-1), Gadd45, and HSP70. Cysteine 39-47 heme oxygenase 1 Homo sapiens 316-332 20385560-6 2010 Covalent modification at two conserved cysteine residues, corresponding to Cys(261) and Cys(273) in HDAC1, coincided with attenuation of histone deacetylase activity, changes in histone H3 and H4 acetylation patterns, derepression of a LEF1.beta-catenin model system, and transcription of HDAC-repressed genes, e.g. heme oxygenase-1 (HO-1), Gadd45, and HSP70. Cysteine 75-78 heme oxygenase 1 Homo sapiens 316-332 20385560-6 2010 Covalent modification at two conserved cysteine residues, corresponding to Cys(261) and Cys(273) in HDAC1, coincided with attenuation of histone deacetylase activity, changes in histone H3 and H4 acetylation patterns, derepression of a LEF1.beta-catenin model system, and transcription of HDAC-repressed genes, e.g. heme oxygenase-1 (HO-1), Gadd45, and HSP70. Cysteine 88-91 heme oxygenase 1 Homo sapiens 316-332 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Heme 43-47 heme oxygenase 1 Homo sapiens 18-22 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 0-16 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 18-22 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Biliverdine 86-96 heme oxygenase 1 Homo sapiens 0-16 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Biliverdine 86-96 heme oxygenase 1 Homo sapiens 18-22 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Bilirubin 97-106 heme oxygenase 1 Homo sapiens 0-16 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Bilirubin 97-106 heme oxygenase 1 Homo sapiens 18-22 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Iron 116-120 heme oxygenase 1 Homo sapiens 0-16 20518085-1 2010 Heme oxygenase-1 (HO-1) system catabolizes heme into three products: carbon monoxide, biliverdin/bilirubin and free iron. Iron 116-120 heme oxygenase 1 Homo sapiens 18-22 20345481-5 2010 When U937 cells were treated with phorbol myristate acetate (PHA) or gamma-interferon, they significantly expressed both HO-1 and Bach1, like primary AML cells. Tetradecanoylphorbol Acetate 34-59 heme oxygenase 1 Homo sapiens 121-125 20523338-0 2010 Neferine inhibits angiotensin II-stimulated proliferation in vascular smooth muscle cells through heme oxygenase-1. neferine 0-8 heme oxygenase 1 Homo sapiens 98-114 20651363-0 2010 Inhibition of heme oxygenase-1 enhances the cytotoxic effect of gemcitabine in urothelial cancer cells. gemcitabine 64-75 heme oxygenase 1 Homo sapiens 14-30 20370320-2 2010 Several clinical and epidemiological studies have been carried out on key enzymes generating and eliminating bilirubin (heme oxygenase-1 and UDP-glucuronosyltransferase UGT1A1, respectively) and their regulation by the AhR. Bilirubin 109-118 heme oxygenase 1 Homo sapiens 120-136 20370564-3 2010 Cell treatment with H(2)O(2) (0.5 mM) resulted in a time- and dose-dependent response of HOX-1 and COX-2 mRNA and protein levels, with ERK1/2, p38-MAPK and MSK1 found to mediate these effects. Hydrogen Peroxide 20-28 heme oxygenase 1 Homo sapiens 89-94 20349048-9 2010 Finally, it was observed that the nitric oxide (NO) anti-inflammatory effects on ICAM-1 expression appear to be indirectly mediated by HO-1 activation, since the inhibition of HO-1 prevented the inhibitory effect of the NO donor (S-nitroso-N-acetylpenicillamine) on LPS-induced ICAM-1 expression. Nitric Oxide 34-46 heme oxygenase 1 Homo sapiens 135-139 21179939-4 2010 We examine here the effects of multiwalled carbon nanotubes of different sizes on monocytic THP-1 cells, with regard to their ability to stimulate increased expression of the HO-1 and GST genes and their ability to produce nuclear translocation of the transcription factor, Nrf2, as well as the release of several pro-inflammatory cytokines and mediators of inflammation. Carbon 43-49 heme oxygenase 1 Homo sapiens 175-179 21179939-5 2010 Our results suggest that long (50 microm) carbon nanotubes (62.5 microg/ml for 4 hours) produce increased nuclear translocation of Nrf2 and increased HO-1 gene expression compared with shorter entangled nanotubes. Carbon 42-48 heme oxygenase 1 Homo sapiens 150-154 20519851-0 2010 Reduction of arsenic-induced cytotoxicity through Nrf2/HO-1 signaling in HepG2 cells. Arsenic 13-20 heme oxygenase 1 Homo sapiens 55-59 20526373-8 2010 Silencing RNA experiments confirmed that HIF-1alpha nuclear translocation was responsible for HO-1, BVR and H-ferritin induction mediated by CS and hypoxia/reoxygenation. Cesium 141-143 heme oxygenase 1 Homo sapiens 94-98 20353835-5 2010 To our knowledge, our results were the first to show that: (a) SIN pretreatment was able to induce HO-1 expression in donor livers in a dose dependent manner; (b) SIN pretreatment protected the liver graft from cold I/R injury; and (c) the protective effect of SIN was, at least in part, mediated by HO-1, as proved by the fact that inhibiting HO-1 activity with zinc protoporphyrin (ZnPP) reduced the protection. sinomenine 63-66 heme oxygenase 1 Homo sapiens 99-103 20353835-5 2010 To our knowledge, our results were the first to show that: (a) SIN pretreatment was able to induce HO-1 expression in donor livers in a dose dependent manner; (b) SIN pretreatment protected the liver graft from cold I/R injury; and (c) the protective effect of SIN was, at least in part, mediated by HO-1, as proved by the fact that inhibiting HO-1 activity with zinc protoporphyrin (ZnPP) reduced the protection. sinomenine 63-66 heme oxygenase 1 Homo sapiens 300-304 20353835-5 2010 To our knowledge, our results were the first to show that: (a) SIN pretreatment was able to induce HO-1 expression in donor livers in a dose dependent manner; (b) SIN pretreatment protected the liver graft from cold I/R injury; and (c) the protective effect of SIN was, at least in part, mediated by HO-1, as proved by the fact that inhibiting HO-1 activity with zinc protoporphyrin (ZnPP) reduced the protection. sinomenine 63-66 heme oxygenase 1 Homo sapiens 300-304 20351094-1 2010 HO-1 (heme oxygenase-1) is an inducible microsomal enzyme that catalyzes the degradation of pro-oxidant heme. Heme 6-10 heme oxygenase 1 Homo sapiens 0-4 20351094-8 2010 Sp1 small interfering RNA and mithramycin A, a Sp1 binding site inhibitor, resulted in loss of the loop formation between the intronic enhancer and the distal HO-1 promoter by the chromosome conformation capture assay. mithramycin A 30-43 heme oxygenase 1 Homo sapiens 159-163 20349048-9 2010 Finally, it was observed that the nitric oxide (NO) anti-inflammatory effects on ICAM-1 expression appear to be indirectly mediated by HO-1 activation, since the inhibition of HO-1 prevented the inhibitory effect of the NO donor (S-nitroso-N-acetylpenicillamine) on LPS-induced ICAM-1 expression. Nitric Oxide 34-46 heme oxygenase 1 Homo sapiens 176-180 20349048-9 2010 Finally, it was observed that the nitric oxide (NO) anti-inflammatory effects on ICAM-1 expression appear to be indirectly mediated by HO-1 activation, since the inhibition of HO-1 prevented the inhibitory effect of the NO donor (S-nitroso-N-acetylpenicillamine) on LPS-induced ICAM-1 expression. S-Nitroso-N-Acetylpenicillamine 230-261 heme oxygenase 1 Homo sapiens 135-139 20349048-9 2010 Finally, it was observed that the nitric oxide (NO) anti-inflammatory effects on ICAM-1 expression appear to be indirectly mediated by HO-1 activation, since the inhibition of HO-1 prevented the inhibitory effect of the NO donor (S-nitroso-N-acetylpenicillamine) on LPS-induced ICAM-1 expression. S-Nitroso-N-Acetylpenicillamine 230-261 heme oxygenase 1 Homo sapiens 176-180 20061555-5 2010 Inhibition of HIV-1 replication was associated with PKC-dependent induction of HO-1, a cytoprotective enzyme known to catabolize heme. Heme 129-133 heme oxygenase 1 Homo sapiens 79-83 20121710-5 2010 However, the patients with short (<24 GT) dinucleotide repeat in the HO-1 gene promoter on either allele had significantly higher prolonged unconjugated hyperbilirubinaemia than the healthy newborns. Dinucleoside Phosphates 45-57 heme oxygenase 1 Homo sapiens 72-76 20127796-7 2010 The effects of miR-196 mimic on Bach1, HMOX1, and HCV RNA, and protein levels were measured by way of quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting, respectively. mir-196 15-22 heme oxygenase 1 Homo sapiens 39-44 20127796-9 2010 miR-196 mimic significantly down-regulated Bach1 and up-regulated HMOX1 gene expression and inhibited HCV expression. mir-196 0-7 heme oxygenase 1 Homo sapiens 66-71 20127796-12 2010 CONCLUSION: miR-196 directly acts on the 3"-UTR of Bach1 messenger RNA and translationally represses the expression of this protein, and up-regulates HMOX1. mir-196 12-19 heme oxygenase 1 Homo sapiens 150-155 20188821-8 2010 These results demonstrate that CSPE-induced ROS generation is mediated through a c-Src/NADPH oxidase/MAPK pathway and in turn initiates the activation of Nrf2 and ultimately induces HO-1 expression in HTSMCs. ros 44-47 heme oxygenase 1 Homo sapiens 182-186 20188822-6 2010 When a competitive HO-1 inhibitor, tin mesoporphyrin, was applied to HepG2 cells or the cells were transfected with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. tin mesoporphyrin 35-52 heme oxygenase 1 Homo sapiens 19-23 20188822-6 2010 When a competitive HO-1 inhibitor, tin mesoporphyrin, was applied to HepG2 cells or the cells were transfected with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. tin mesoporphyrin 35-52 heme oxygenase 1 Homo sapiens 126-130 20188822-6 2010 When a competitive HO-1 inhibitor, tin mesoporphyrin, was applied to HepG2 cells or the cells were transfected with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. Polychlorinated Dibenzodioxins 184-188 heme oxygenase 1 Homo sapiens 19-23 20188822-6 2010 When a competitive HO-1 inhibitor, tin mesoporphyrin, was applied to HepG2 cells or the cells were transfected with siRNA for HO-1 there was a partial restoration of the inhibition of TCDD-mediated induction of CYP1A1 catalytic activity. Polychlorinated Dibenzodioxins 184-188 heme oxygenase 1 Homo sapiens 126-130 20188822-8 2010 On the other hand, cobalt protoporphyrin increased HO-1 mRNA, with a concomitant decrease in CYP1A1 activity, without affecting CYP1A1 mRNA, which was reversed by HO-1 siRNA transfection. cobaltiprotoporphyrin 19-40 heme oxygenase 1 Homo sapiens 51-55 20404253-8 2010 In isolated mitochondria, mitochondrial permeability transition was inhibited by HO-1 in a carbon monoxide (CO)-dependent manner and was recapitulated by the CO donor tricarbonylchloro(glycinato)ruthenium(II) (CORM-3). Carbon Monoxide 91-106 heme oxygenase 1 Homo sapiens 81-85 20404253-8 2010 In isolated mitochondria, mitochondrial permeability transition was inhibited by HO-1 in a carbon monoxide (CO)-dependent manner and was recapitulated by the CO donor tricarbonylchloro(glycinato)ruthenium(II) (CORM-3). Carbon Monoxide 108-111 heme oxygenase 1 Homo sapiens 81-85 20404253-8 2010 In isolated mitochondria, mitochondrial permeability transition was inhibited by HO-1 in a carbon monoxide (CO)-dependent manner and was recapitulated by the CO donor tricarbonylchloro(glycinato)ruthenium(II) (CORM-3). tricarbonylchloro(glycinato)ruthenium(II) 167-207 heme oxygenase 1 Homo sapiens 81-85 19924377-6 2010 On addition of a cobalt compound, the resultant induction of HO-1 decreases adipogenesis. Cobalt 17-23 heme oxygenase 1 Homo sapiens 61-65 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Iron 95-99 heme oxygenase 1 Homo sapiens 37-40 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Reactive Oxygen Species 113-136 heme oxygenase 1 Homo sapiens 37-40 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Reactive Oxygen Species 138-141 heme oxygenase 1 Homo sapiens 37-40 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Abscisic Acid 144-157 heme oxygenase 1 Homo sapiens 37-40 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Abscisic Acid 159-162 heme oxygenase 1 Homo sapiens 37-40 20378668-7 2010 The transcriptional up-regulation of HO1 in plants responds to many agents, such as light, UV, iron deprivation, reactive oxygen species (ROS), abscisic acid (ABA), and haematin. Hemin 169-177 heme oxygenase 1 Homo sapiens 37-40 20336713-6 2010 The attenuation of iron-induced augmentation of heme oxygenase-1 was also confirmed in HepG2 cells expressing the core protein. Iron 19-23 heme oxygenase 1 Homo sapiens 48-64 20061555-6 2010 Pretreatment with the PKC inhibitor Go 6976 not only substantially inhibited LPS-mediated induction of HO-1 but also attenuated LPS-induced suppression of HIV replication. Go 6976 36-43 heme oxygenase 1 Homo sapiens 103-107 20061555-8 2010 Specificity of HO-1 was confirmed by substantial reversal of LPS-induced viral replication by pretreatment of cells with SnPP IX, an inhibitor of HO-1 enzyme activity. tin protoporphyrin IX 121-128 heme oxygenase 1 Homo sapiens 15-19 20061555-8 2010 Specificity of HO-1 was confirmed by substantial reversal of LPS-induced viral replication by pretreatment of cells with SnPP IX, an inhibitor of HO-1 enzyme activity. tin protoporphyrin IX 121-128 heme oxygenase 1 Homo sapiens 146-150 20110250-6 2010 HO-1 was induced by cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 20-44 heme oxygenase 1 Homo sapiens 0-4 20106603-4 2010 In this context, it has been shown that the rate limiting enzyme heme oxygenase I (HO-1), responsible for the catabolism of the free heme in the body, is an important resistance factor in malaria and is also important in the physiopathology of haemolytic diseases. Heme 65-69 heme oxygenase 1 Homo sapiens 83-87 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Mesalamine 59-80 heme oxygenase 1 Homo sapiens 210-226 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Mesalamine 59-80 heme oxygenase 1 Homo sapiens 228-232 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Mesalamine 82-87 heme oxygenase 1 Homo sapiens 210-226 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Mesalamine 82-87 heme oxygenase 1 Homo sapiens 228-232 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Aminosalicylic Acid 123-138 heme oxygenase 1 Homo sapiens 210-226 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Aminosalicylic Acid 123-138 heme oxygenase 1 Homo sapiens 228-232 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Sulfasalazine 160-173 heme oxygenase 1 Homo sapiens 210-226 20631420-2 2010 Recent in vivo and in vitro studies have demonstrated that 5-aminosalicylic acid (5-ASA), regarded as the active moiety in aminosalicylate preparations such as sulfasalazine, can induce the heat shock protein, heme oxygenase-1 (HO-1) and up-regulate HO enzyme activity in the colon. Sulfasalazine 160-173 heme oxygenase 1 Homo sapiens 228-232 20631420-3 2010 As HO-1 can produce endogenous anti-oxidant and anti-inflammatory moieties such as bilirubin and carbon monoxide (CO), these findings suggest a novel mechanism of action for aminosalicylates, acting as anti-colitic agents through the up-regulation of HO-1 enzyme expression and activity. Bilirubin 83-92 heme oxygenase 1 Homo sapiens 3-7 20631420-3 2010 As HO-1 can produce endogenous anti-oxidant and anti-inflammatory moieties such as bilirubin and carbon monoxide (CO), these findings suggest a novel mechanism of action for aminosalicylates, acting as anti-colitic agents through the up-regulation of HO-1 enzyme expression and activity. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 3-7 20631420-3 2010 As HO-1 can produce endogenous anti-oxidant and anti-inflammatory moieties such as bilirubin and carbon monoxide (CO), these findings suggest a novel mechanism of action for aminosalicylates, acting as anti-colitic agents through the up-regulation of HO-1 enzyme expression and activity. Carbon Monoxide 114-116 heme oxygenase 1 Homo sapiens 3-7 20631420-3 2010 As HO-1 can produce endogenous anti-oxidant and anti-inflammatory moieties such as bilirubin and carbon monoxide (CO), these findings suggest a novel mechanism of action for aminosalicylates, acting as anti-colitic agents through the up-regulation of HO-1 enzyme expression and activity. Carbon Monoxide 114-116 heme oxygenase 1 Homo sapiens 251-255 20110250-6 2010 HO-1 was induced by cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 46-50 heme oxygenase 1 Homo sapiens 0-4 20110250-11 2010 RESULTS: Induction of HO-1 by CoPP in the presence of IL-1beta decreased the expression of MMP-1 and -3, and MMP activity. cobaltiprotoporphyrin 30-34 heme oxygenase 1 Homo sapiens 22-26 20083128-0 2010 Arsenic promotes angiogenesis in vitro via a heme oxygenase-1-dependent mechanism. Arsenic 0-7 heme oxygenase 1 Homo sapiens 45-61 20083128-4 2010 The present study investigates the role of heme oxygenase-1 (HO-1) in sodium arsenite-mediated angiogenesis in vitro. sodium arsenite 70-85 heme oxygenase 1 Homo sapiens 43-59 20083128-4 2010 The present study investigates the role of heme oxygenase-1 (HO-1) in sodium arsenite-mediated angiogenesis in vitro. sodium arsenite 70-85 heme oxygenase 1 Homo sapiens 61-65 20083128-8 2010 Arsenite induced HO-1 mRNA and protein expression. arsenite 0-8 heme oxygenase 1 Homo sapiens 17-21 20083128-9 2010 Knock down of HO-1 expression decreased arsenite-induced VEGF expression, cell migration, and tube formation. arsenite 40-48 heme oxygenase 1 Homo sapiens 14-18 20083128-10 2010 We showed that arsenite promoted dissociation of Bach1 (a transcriptional repressor) from the HO-1 enhancers and increased Nrf2 binding to these elements. arsenite 15-23 heme oxygenase 1 Homo sapiens 94-98 20083128-11 2010 Site directed mutagenesis assay identified that Bach1 cysteine residues 557 and 574 were essential for the induction of HO-1 gene in response to arsenite. Cysteine 54-62 heme oxygenase 1 Homo sapiens 120-124 20083128-11 2010 Site directed mutagenesis assay identified that Bach1 cysteine residues 557 and 574 were essential for the induction of HO-1 gene in response to arsenite. arsenite 145-153 heme oxygenase 1 Homo sapiens 120-124 20083128-12 2010 These findings demonstrate a role for HO-1 in arsenite-mediated angiogenesis in vitro. arsenite 46-54 heme oxygenase 1 Homo sapiens 38-42 20368435-7 2010 Our results show that compounds containing alpha-beta unsaturated carbonyls, sulfhydryl reactive metals, and isothiocyanates are strong activators of Nrf2 in a reporter assay system and in primary human CLL based on increased expression of the Nrf2 target heme oxygenase-1. alpha-beta unsaturated carbonyls 43-75 heme oxygenase 1 Homo sapiens 256-272 20368435-7 2010 Our results show that compounds containing alpha-beta unsaturated carbonyls, sulfhydryl reactive metals, and isothiocyanates are strong activators of Nrf2 in a reporter assay system and in primary human CLL based on increased expression of the Nrf2 target heme oxygenase-1. Isothiocyanates 109-124 heme oxygenase 1 Homo sapiens 256-272 19890094-5 2010 The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. Carbon Monoxide 126-141 heme oxygenase 1 Homo sapiens 39-43 20083195-8 2010 Furthermore, celecoxib-induced HO-1 was inhibited by dominant-negative Nrf2. Celecoxib 13-22 heme oxygenase 1 Homo sapiens 31-35 20083195-9 2010 The functional significance of HO-1 induction was revealed by celecoxib-mediated inhibition of VCAM-1 expression, a response reversed by the HO-1 antagonist zinc protoporphyrin. Celecoxib 62-71 heme oxygenase 1 Homo sapiens 31-35 20083195-9 2010 The functional significance of HO-1 induction was revealed by celecoxib-mediated inhibition of VCAM-1 expression, a response reversed by the HO-1 antagonist zinc protoporphyrin. Celecoxib 62-71 heme oxygenase 1 Homo sapiens 141-145 20083195-9 2010 The functional significance of HO-1 induction was revealed by celecoxib-mediated inhibition of VCAM-1 expression, a response reversed by the HO-1 antagonist zinc protoporphyrin. zinc protoporphyrin 157-176 heme oxygenase 1 Homo sapiens 31-35 20083195-9 2010 The functional significance of HO-1 induction was revealed by celecoxib-mediated inhibition of VCAM-1 expression, a response reversed by the HO-1 antagonist zinc protoporphyrin. zinc protoporphyrin 157-176 heme oxygenase 1 Homo sapiens 141-145 20083195-10 2010 HO-1 induction provides a molecular mechanism for clinical observations indicating relative freedom from atherothrombotic complications in patients taking celecoxib compared to other NSAIDs with comparable anti-inflammatory activity. Celecoxib 155-164 heme oxygenase 1 Homo sapiens 0-4 20083195-0 2010 Celecoxib activates PI-3K/Akt and mitochondrial redox signaling to enhance heme oxygenase-1-mediated anti-inflammatory activity in vascular endothelium. Celecoxib 0-9 heme oxygenase 1 Homo sapiens 75-91 20083195-3 2010 This led us to the hypothesis that celecoxib induces the vasculoprotective enzyme heme oxygenase-1 (HO-1). Celecoxib 35-44 heme oxygenase 1 Homo sapiens 82-98 20083195-3 2010 This led us to the hypothesis that celecoxib induces the vasculoprotective enzyme heme oxygenase-1 (HO-1). Celecoxib 35-44 heme oxygenase 1 Homo sapiens 100-104 20083195-4 2010 In human umbilical vein and aortic endothelial cells, 24-48 h treatment with celecoxib induced HO-1 mRNA and protein expression and increased HO-1 enzyme activity. Celecoxib 77-86 heme oxygenase 1 Homo sapiens 95-99 20083195-4 2010 In human umbilical vein and aortic endothelial cells, 24-48 h treatment with celecoxib induced HO-1 mRNA and protein expression and increased HO-1 enzyme activity. Celecoxib 77-86 heme oxygenase 1 Homo sapiens 142-146 20223678-5 2010 It was shown that inhibition of HO-1 activity by zinc protoporphyrin IX increased PDT-induced cytotoxicity in a dose-dependent manner. zinc protoporphyrin 49-71 heme oxygenase 1 Homo sapiens 32-36 20223678-9 2010 This is supported by the fact that during PDT ferritin is readily up-regulated, able to bind excess iron formed by the HO-1 action. Iron 100-104 heme oxygenase 1 Homo sapiens 119-123 19890094-5 2010 The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. mesoporphyrin IX 222-238 heme oxygenase 1 Homo sapiens 111-115 19890094-5 2010 The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. Bilirubin 147-156 heme oxygenase 1 Homo sapiens 39-43 19890094-5 2010 The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. mesoporphyrin IX 222-238 heme oxygenase 1 Homo sapiens 39-43 19890094-5 2010 The protective effect was specific for HO-1 because it was reproduced on administration of the end products of HO-1 activity, carbon monoxide, and bilirubin, and prevented by the pharmacologic inhibition of HO-1 using tin mesoporphyrin IX. mesoporphyrin IX 222-238 heme oxygenase 1 Homo sapiens 111-115 20470258-0 2010 Heme oxygenase-1 mediates nicotine- and lipopolysaccharide-induced expression of cyclooxygenase-2 and inducible nitric oxide synthase in human periodontal ligament cells. Nicotine 26-34 heme oxygenase 1 Homo sapiens 0-16 19885829-5 2010 Following exposure of cells to EA, heme oxygenase-1, a marker protein of oxidative stress, was strongly induced. Ethacrynic Acid 31-33 heme oxygenase 1 Homo sapiens 35-51 20470258-2 2010 This study aimed to identify the effects of HO-1 on the proinflammatory mediators activated by nicotine and lipopolysaccharide (LPS) stimulation in human periodontal ligament (PDL) cells. Nicotine 95-103 heme oxygenase 1 Homo sapiens 44-48 20470258-5 2010 RESULTS: Lipopolysaccharide and nicotine synergistically induced the production of NO and PGE(2) and increased the protein expression of iNOS, COX-2 and HO-1. Nicotine 32-40 heme oxygenase 1 Homo sapiens 153-157 20470258-6 2010 Treatment with an HO-1 inhibitor and HO-1 small interfering RNAs blocked the LPS- and nicotine-stimulated NO and PGE(2) release as well as the expression of iNOS and COX-2. Nicotine 86-94 heme oxygenase 1 Homo sapiens 18-22 20470258-6 2010 Treatment with an HO-1 inhibitor and HO-1 small interfering RNAs blocked the LPS- and nicotine-stimulated NO and PGE(2) release as well as the expression of iNOS and COX-2. Nicotine 86-94 heme oxygenase 1 Homo sapiens 37-41 20470258-6 2010 Treatment with an HO-1 inhibitor and HO-1 small interfering RNAs blocked the LPS- and nicotine-stimulated NO and PGE(2) release as well as the expression of iNOS and COX-2. Dinoprostone 113-119 heme oxygenase 1 Homo sapiens 18-22 20470258-6 2010 Treatment with an HO-1 inhibitor and HO-1 small interfering RNAs blocked the LPS- and nicotine-stimulated NO and PGE(2) release as well as the expression of iNOS and COX-2. Dinoprostone 113-119 heme oxygenase 1 Homo sapiens 37-41 20470258-7 2010 CONCLUSION: Our data suggest that the nicotine- and LPS-induced inflammatory effects on PDL cells may act through a novel mechanism involving the action of HO-1. Nicotine 38-46 heme oxygenase 1 Homo sapiens 156-160 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Cholesterol 54-65 heme oxygenase 1 Homo sapiens 0-16 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Cholesterol 54-65 heme oxygenase 1 Homo sapiens 18-22 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Oxysterols 69-79 heme oxygenase 1 Homo sapiens 0-16 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Oxysterols 69-79 heme oxygenase 1 Homo sapiens 18-22 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Oxysterols 69-78 heme oxygenase 1 Homo sapiens 0-16 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Oxysterols 69-78 heme oxygenase 1 Homo sapiens 18-22 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Cholesterol 273-284 heme oxygenase 1 Homo sapiens 0-16 18597895-2 2010 Heme oxygenase-1 (HO-1) stimulates oxidation of glial cholesterol to oxysterols, and increased oxysterol concentrations may protect neural tissues by activation of liver X receptor-beta (LXR-beta), which induces transcription of genes associated with reduction of cellular cholesterol concentrations and decrease of Abeta formation. Cholesterol 273-284 heme oxygenase 1 Homo sapiens 18-22 18597895-3 2010 Underexpression of HO-1 in concert with underexpression of LXR-beta would result in increased cholesterol accumulation, induction of Abeta production, and increased AD risk. Cholesterol 94-105 heme oxygenase 1 Homo sapiens 19-23 20131233-12 2010 Cilostazol- and CoPP-induced HO-1 expression was diminished in cells transfected with Nrf2 siRNA. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 29-33 20170153-0 2010 Hesperidin upregulates heme oxygenase-1 to attenuate hydrogen peroxide-induced cell damage in hepatic L02 cells. Hydrogen Peroxide 53-70 heme oxygenase 1 Homo sapiens 23-39 20170153-4 2010 Real-time quantitative polymerase chain reaction, Western blot, and enzyme activity assay demonstrated that hesperidin upregulated heme oxygenase-1 (HO-1) expression to protect hepatocytes against oxidative stress. Hesperidin 108-118 heme oxygenase 1 Homo sapiens 131-147 20170153-4 2010 Real-time quantitative polymerase chain reaction, Western blot, and enzyme activity assay demonstrated that hesperidin upregulated heme oxygenase-1 (HO-1) expression to protect hepatocytes against oxidative stress. Hesperidin 108-118 heme oxygenase 1 Homo sapiens 149-153 20170153-7 2010 Besides, ERK1/2 inhibitor significantly inhibited hesperidin-mediated HO-1 upregulation and Nrf2 nuclear translocation. Hesperidin 50-60 heme oxygenase 1 Homo sapiens 70-74 20170153-8 2010 Taken together, the above findings suggested that hesperidin augmented cellular antioxidant defense capacity through the induction of HO-1 via ERK/Nrf2 signaling. Hesperidin 50-60 heme oxygenase 1 Homo sapiens 134-138 20118244-1 2010 Heme oxygenases (HOs) -1 and -2 catalyze the breakdown of heme to release carbon monoxide, biliverdin, and ferrous iron, which may preserve cell function during oxidative stress. Heme 58-62 heme oxygenase 1 Homo sapiens 0-31 20118244-1 2010 Heme oxygenases (HOs) -1 and -2 catalyze the breakdown of heme to release carbon monoxide, biliverdin, and ferrous iron, which may preserve cell function during oxidative stress. Carbon Monoxide 74-89 heme oxygenase 1 Homo sapiens 0-31 20118244-1 2010 Heme oxygenases (HOs) -1 and -2 catalyze the breakdown of heme to release carbon monoxide, biliverdin, and ferrous iron, which may preserve cell function during oxidative stress. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 0-31 20118244-1 2010 Heme oxygenases (HOs) -1 and -2 catalyze the breakdown of heme to release carbon monoxide, biliverdin, and ferrous iron, which may preserve cell function during oxidative stress. Iron 115-119 heme oxygenase 1 Homo sapiens 0-31 20131233-0 2010 Cilostazol enhances apoptosis of synovial cells from rheumatoid arthritis patients with inhibition of cytokine formation via Nrf2-linked heme oxygenase 1 induction. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 137-153 20131233-6 2010 Cilostazol (10 microM) and cobalt protoporphyrin IX (CoPP) increased HO-1 messenger RNA and protein expression. Cilostazol 0-10 heme oxygenase 1 Homo sapiens 69-73 20131233-6 2010 Cilostazol (10 microM) and cobalt protoporphyrin IX (CoPP) increased HO-1 messenger RNA and protein expression. cobaltiprotoporphyrin 27-51 heme oxygenase 1 Homo sapiens 69-73 20131233-12 2010 Cilostazol- and CoPP-induced HO-1 expression was diminished in cells transfected with Nrf2 siRNA. cobaltiprotoporphyrin 16-20 heme oxygenase 1 Homo sapiens 29-33 20131233-6 2010 Cilostazol (10 microM) and cobalt protoporphyrin IX (CoPP) increased HO-1 messenger RNA and protein expression. cobaltiprotoporphyrin 53-57 heme oxygenase 1 Homo sapiens 69-73 20131233-13 2010 CONCLUSION: Cilostazol suppressed proliferation of synovial cells from RA patients by enhancing apoptosis, and also inhibited cytokine production via mediation of cAMP-dependent protein kinase activation-coupled Nrf2-linked HO-1 expression. Cilostazol 12-22 heme oxygenase 1 Homo sapiens 224-228 20131233-7 2010 These effects were suppressed by zinc protoporphyrin IX (ZnPP), an HO-1 inhibitor. zinc protoporphyrin 33-55 heme oxygenase 1 Homo sapiens 67-71 20131233-7 2010 These effects were suppressed by zinc protoporphyrin IX (ZnPP), an HO-1 inhibitor. zinc protoporphyrin 57-61 heme oxygenase 1 Homo sapiens 67-71 20131233-13 2010 CONCLUSION: Cilostazol suppressed proliferation of synovial cells from RA patients by enhancing apoptosis, and also inhibited cytokine production via mediation of cAMP-dependent protein kinase activation-coupled Nrf2-linked HO-1 expression. Cyclic AMP 163-167 heme oxygenase 1 Homo sapiens 224-228 20091384-1 2010 Heme oxygenase (HO)-1, a heme-degrading enzyme inducible by various stimuli, plays a key role in the regulation of inflammatory response in monocytes/macrophages. Heme 25-29 heme oxygenase 1 Homo sapiens 0-21 20124108-5 2010 Overexpression of heme oxygenase 1 and its gaseous product, carbon monoxide, decreased PAR-2-stimulated sVEGFR-1 release from human umbilical vein endothelial cells. Carbon Monoxide 60-75 heme oxygenase 1 Homo sapiens 18-34 20124108-6 2010 Simvastatin, which upregulates heme oxygenase 1, also suppressed PAR-2-mediated sVEGFR-1 release. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 31-47 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Heme 139-143 heme oxygenase 1 Homo sapiens 0-16 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Heme 139-143 heme oxygenase 1 Homo sapiens 18-22 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 0-16 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 18-22 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Biliverdine 169-179 heme oxygenase 1 Homo sapiens 0-16 20020468-1 2010 Heme-oxygenase-1 (HO-1), an important enzyme involved in vascular disease, transplantation, and inflammation, catalyzes the degradation of heme into carbon monoxide and biliverdin. Biliverdine 169-179 heme oxygenase 1 Homo sapiens 18-22 20020468-6 2010 Furthermore, downstream products of HO-1, bilirubin, carbon monoxide, and iron, are involved in the inhibitory action of HO-1. Bilirubin 42-51 heme oxygenase 1 Homo sapiens 36-40 20171361-0 2010 Simvastatin promotes odontoblastic differentiation and expression of angiogenic factors via heme oxygenase-1 in primary cultured human dental pulp cells. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 92-108 20020468-6 2010 Furthermore, downstream products of HO-1, bilirubin, carbon monoxide, and iron, are involved in the inhibitory action of HO-1. Bilirubin 42-51 heme oxygenase 1 Homo sapiens 121-125 20171361-7 2010 The inducing effect of simvastatin on odontoblastic differentiation and angiogenesis was nullified by an HO-1 inhibitor and a carbon monoxide (CO) scavenger. Simvastatin 23-34 heme oxygenase 1 Homo sapiens 105-109 20020468-6 2010 Furthermore, downstream products of HO-1, bilirubin, carbon monoxide, and iron, are involved in the inhibitory action of HO-1. Carbon Monoxide 53-68 heme oxygenase 1 Homo sapiens 121-125 20171361-8 2010 CONCLUSIONS: These results suggested that simvastatin exerts its odontoblastic differentiation and angiogenesis-inducing effects in HDPCs through a mechanism that involves the action of HO-1 and its product CO. Simvastatin 42-53 heme oxygenase 1 Homo sapiens 186-190 20020468-6 2010 Furthermore, downstream products of HO-1, bilirubin, carbon monoxide, and iron, are involved in the inhibitory action of HO-1. Iron 74-78 heme oxygenase 1 Homo sapiens 121-125 20020468-8 2010 In addition, endogenous PPARgamma ligand, 15-deoxy-Delta(12,14)-prostaglandin-J2 (15d-PGJ2) markedly increased both mRNA and protein levels of HO-1 in osteoblasts via PI3K-Akt and MAPK pathways. 15-deoxy-delta( 42-57 heme oxygenase 1 Homo sapiens 143-147 19917515-0 2010 Structural characterization of human heme oxygenase-1 in complex with azole-based inhibitors. Azoles 70-75 heme oxygenase 1 Homo sapiens 37-53 20020468-8 2010 In addition, endogenous PPARgamma ligand, 15-deoxy-Delta(12,14)-prostaglandin-J2 (15d-PGJ2) markedly increased both mRNA and protein levels of HO-1 in osteoblasts via PI3K-Akt and MAPK pathways. 9-deoxy-delta-9-prostaglandin D2 63-80 heme oxygenase 1 Homo sapiens 143-147 20333281-0 2010 Possible role of heme oxygenase-1 and prostaglandins in the pathogenesis of cerebral malaria: heme oxygenase-1 induction by prostaglandin D(2) and metabolite by a human astrocyte cell line. Prostaglandins 38-52 heme oxygenase 1 Homo sapiens 94-110 20333281-0 2010 Possible role of heme oxygenase-1 and prostaglandins in the pathogenesis of cerebral malaria: heme oxygenase-1 induction by prostaglandin D(2) and metabolite by a human astrocyte cell line. Prostaglandins D 124-139 heme oxygenase 1 Homo sapiens 94-110 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Heme 48-52 heme oxygenase 1 Homo sapiens 0-16 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Heme 48-52 heme oxygenase 1 Homo sapiens 18-22 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Iron 82-86 heme oxygenase 1 Homo sapiens 0-16 20217200-7 2010 On the other hand, heme oxygenase-1 (HO-1) and inducible nitric oxide synthase (iNOS) expression were significantly increased by exposure to both ammonia and proinflammatory mediators and although modest, these effects were additive suggestive of a synergistic mechanism. Ammonia 146-153 heme oxygenase 1 Homo sapiens 19-35 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Iron 82-86 heme oxygenase 1 Homo sapiens 18-22 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 0-16 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 18-22 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 0-16 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Biliverdine 109-119 heme oxygenase 1 Homo sapiens 18-22 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Bilirubin 120-129 heme oxygenase 1 Homo sapiens 0-16 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Bilirubin 120-129 heme oxygenase 1 Homo sapiens 18-22 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Iron 149-153 heme oxygenase 1 Homo sapiens 0-16 20333281-4 2010 Heme oxygenase-1 (HO-1) is catalyzing enzyme in heme breakdown process to release iron, carbon monoxide, and biliverdin/bilirubin, and may influence iron supply to the P. falciparum parasites. Iron 149-153 heme oxygenase 1 Homo sapiens 18-22 20203419-0 2010 4-Methylcatechol-induced heme oxygenase-1 exerts a protective effect against oxidative stress in cultured neural stem/progenitor cells via PI3 kinase/Akt pathway. 4-methylcatechol 0-16 heme oxygenase 1 Homo sapiens 25-41 19961840-0 2010 Senkyunolides reduce hydrogen peroxide-induced oxidative damage in human liver HepG2 cells via induction of heme oxygenase-1. 3-N-butyl-4,5-dihydrophthalide 0-13 heme oxygenase 1 Homo sapiens 108-124 19961840-0 2010 Senkyunolides reduce hydrogen peroxide-induced oxidative damage in human liver HepG2 cells via induction of heme oxygenase-1. Hydrogen Peroxide 21-38 heme oxygenase 1 Homo sapiens 108-124 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Heme 122-126 heme oxygenase 1 Homo sapiens 140-144 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Biliverdine 221-231 heme oxygenase 1 Homo sapiens 140-144 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Carbon Monoxide 233-248 heme oxygenase 1 Homo sapiens 122-138 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Carbon Monoxide 233-248 heme oxygenase 1 Homo sapiens 140-144 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Iron 258-262 heme oxygenase 1 Homo sapiens 122-138 19961840-2 2010 To delineate the underlying molecular mechanisms, we recently found that Rhizoma Chuanxiong extract significantly induced heme oxygenase-1 (HO-1), an enzyme that degrades intracellular heme into three bioactive products: biliverdin, carbon monoxide and free iron. Iron 258-262 heme oxygenase 1 Homo sapiens 140-144 19961840-9 2010 Thus, this study demonstrated that senkyunolide-H and -I attenuated oxidative damage via activation of HO-1 pathway. 3-N-butyl-4,5-dihydrophthalide 35-47 heme oxygenase 1 Homo sapiens 103-107 19965783-2 2010 Tissues from sickle patients express heme oxygenase-1 (HO-1), the enzyme that degrades free heme/hemoglobin to the signaling molecule carbon monoxide, and the antioxidants biliverdin/bilirubin. Carbon Monoxide 134-149 heme oxygenase 1 Homo sapiens 37-53 19965783-2 2010 Tissues from sickle patients express heme oxygenase-1 (HO-1), the enzyme that degrades free heme/hemoglobin to the signaling molecule carbon monoxide, and the antioxidants biliverdin/bilirubin. Carbon Monoxide 134-149 heme oxygenase 1 Homo sapiens 55-59 19965783-8 2010 HO-1 induction, exogenous biliverdin, or carbon monoxide markedly decreased adhesion of sickle blood to the endothelium, and sickle red cells partially inhibited relaxation mediated by carbon monoxide in isolated aortas. Carbon Monoxide 185-200 heme oxygenase 1 Homo sapiens 0-4 20203419-1 2010 4-Methylcatechol (4MC), a stimulator of the synthesis of neurotrophin family members in various cells, was able to up-regulate the expression of heme oxygenase (HO)-1, a redox-sensitive inducible stress protein, in neural stem/progenitor cells (NS/PCs). 4-methylcatechol 0-16 heme oxygenase 1 Homo sapiens 145-166 20203419-1 2010 4-Methylcatechol (4MC), a stimulator of the synthesis of neurotrophin family members in various cells, was able to up-regulate the expression of heme oxygenase (HO)-1, a redox-sensitive inducible stress protein, in neural stem/progenitor cells (NS/PCs). 4-methylcatechol 18-21 heme oxygenase 1 Homo sapiens 145-166 20203419-2 2010 RT-PCR analysis showed that 4MC induced HO-1 mRNA expression in a dose- and a time-dependent manner. 4-methylcatechol 28-31 heme oxygenase 1 Homo sapiens 40-44 20203419-6 2010 Pretreatment of cultures with a selective inhibitor of PI3 kinase (PI3K)/Akt, but not with one of MAPK/ERK, inhibited both the 4MCinduced HO-1 expression and neuroprotective effect, demonstrating that PI3K/Akt signaling pathway played a significant role in 4MC-induced HO-1 induction and neuroprotection. 4-methylcatechol 127-130 heme oxygenase 1 Homo sapiens 138-142 20203419-6 2010 Pretreatment of cultures with a selective inhibitor of PI3 kinase (PI3K)/Akt, but not with one of MAPK/ERK, inhibited both the 4MCinduced HO-1 expression and neuroprotective effect, demonstrating that PI3K/Akt signaling pathway played a significant role in 4MC-induced HO-1 induction and neuroprotection. 4-methylcatechol 127-130 heme oxygenase 1 Homo sapiens 269-273 20203419-7 2010 Taken together, our results suggest that 4MC activates the expression of HO-1 through the PI3K/Akt signaling pathway and that the HO-1 protein inhibits the death of NS/PCs induced by oxidative stress. 4-methylcatechol 41-44 heme oxygenase 1 Homo sapiens 73-77 19956091-1 2010 Heme oxygenase (HO)-1 degrades heme and protects against oxidative stress, but it has not been pharmacologically induced in humans. Heme 31-35 heme oxygenase 1 Homo sapiens 0-21 19956091-2 2010 In this randomized study of 10 healthy volunteers, hemin (3 mg/kg intravenously in 25% albumin) was shown to increase plasma HO-1 protein concentration four- to fivefold and HO-1 activity ~15-fold relative to baseline at 24 and 48 h (placebo -56.41 +/- 6.31 (baseline), 69.79 +/- 13.00 (24 h), 77.44 +/- 10.62 (48 h) vs. hemin -71.70 +/- 9.20 (baseline), 1,126.20 +/- 293.30 (24 h), 1,192.20 +/- 333.30 (48 h)) in four of five subjects as compared with albumin alone (P </= 0.03). Hemin 51-56 heme oxygenase 1 Homo sapiens 125-129 19956091-2 2010 In this randomized study of 10 healthy volunteers, hemin (3 mg/kg intravenously in 25% albumin) was shown to increase plasma HO-1 protein concentration four- to fivefold and HO-1 activity ~15-fold relative to baseline at 24 and 48 h (placebo -56.41 +/- 6.31 (baseline), 69.79 +/- 13.00 (24 h), 77.44 +/- 10.62 (48 h) vs. hemin -71.70 +/- 9.20 (baseline), 1,126.20 +/- 293.30 (24 h), 1,192.20 +/- 333.30 (48 h)) in four of five subjects as compared with albumin alone (P </= 0.03). Hemin 51-56 heme oxygenase 1 Homo sapiens 174-178 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Heme 117-121 heme oxygenase 1 Homo sapiens 0-16 19558496-6 2010 Induction of Nrf2 due to SFN was accompanied by an increase in mRNA and protein levels of NADPH quinone oxidoreductase 1, heme oxygenase 1 and gamma-glutamylcysteine-synthetase. sulforaphane 25-28 heme oxygenase 1 Homo sapiens 122-138 19797297-1 2010 HO-1 is the only inducible one of three isoenzymes that catalyzes the oxidative degradation of heme. Heme 95-99 heme oxygenase 1 Homo sapiens 0-4 19797297-7 2010 HO-1 modulation by SnPP treatment of T(regs) or PGJ(2) treatment of CD4(+)CD25(-) T cells neither suppressed nor induced immune-modulatory function in these cells, respectively, as measured by responder-cell proliferation and/or IL-2 production. 2-(ETHOXYMETHYL)-4-(4-FLUOROPHENYL)-3-[2-(2-HYDROXYPHENOXY)PYRIMIDIN-4-YL]ISOXAZOL-5(2H)-ONE 48-51 heme oxygenase 1 Homo sapiens 0-4 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Heme 117-121 heme oxygenase 1 Homo sapiens 18-22 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 0-16 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 18-22 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 0-16 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 18-22 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Iron 147-151 heme oxygenase 1 Homo sapiens 0-16 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Iron 147-151 heme oxygenase 1 Homo sapiens 18-22 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Bilirubin 156-165 heme oxygenase 1 Homo sapiens 0-16 19925812-1 2010 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron and bilirubin. Bilirubin 156-165 heme oxygenase 1 Homo sapiens 18-22 19925812-13 2010 These results indicate that the downstream products of HO-1, i.e. bilirubin and CO, modulate BDNF and GDNF expression in neuron and astrocyte. Bilirubin 66-75 heme oxygenase 1 Homo sapiens 55-59 19937094-0 2010 Kaempferol suppresses cisplatin-induced apoptosis via inductions of heme oxygenase-1 and glutamate-cysteine ligase catalytic subunit in HEI-OC1 cell. kaempferol 0-10 heme oxygenase 1 Homo sapiens 68-84 19937094-0 2010 Kaempferol suppresses cisplatin-induced apoptosis via inductions of heme oxygenase-1 and glutamate-cysteine ligase catalytic subunit in HEI-OC1 cell. Cisplatin 22-31 heme oxygenase 1 Homo sapiens 68-84 19937094-7 2010 Kaempferol-induced HO-1 expression protected against cell death though the c-Jun N-terminal kinase (JNK) pathway and by the aid of Nrf2 translocation. kaempferol 0-10 heme oxygenase 1 Homo sapiens 19-23 19937094-10 2010 CONCLUSION: The expression of HO-1 by kaempferol inhibits cisplatin-induced apoptosis in HEI-OC1 cells, and the mechanism of protective effect is also associated with its inductive effect of GCLC expression. kaempferol 38-48 heme oxygenase 1 Homo sapiens 30-34 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. rottlerin 0-9 heme oxygenase 1 Homo sapiens 18-34 19937094-10 2010 CONCLUSION: The expression of HO-1 by kaempferol inhibits cisplatin-induced apoptosis in HEI-OC1 cells, and the mechanism of protective effect is also associated with its inductive effect of GCLC expression. Cisplatin 58-67 heme oxygenase 1 Homo sapiens 30-34 20656217-4 2010 Heme oxygenase 1 (HO-1) protein expression is specifically increased in lesions with a vulnerable plaque phenotype resembling TCFAs and correlates with a rise in expression levels of intimal proinflammatory markers. tcfas 126-131 heme oxygenase 1 Homo sapiens 0-16 20656217-4 2010 Heme oxygenase 1 (HO-1) protein expression is specifically increased in lesions with a vulnerable plaque phenotype resembling TCFAs and correlates with a rise in expression levels of intimal proinflammatory markers. tcfas 126-131 heme oxygenase 1 Homo sapiens 18-22 21081219-6 2010 In the past, attempts to avoid nitrate-induced side effects have focused on administration schedules that would allow a "nitrate-free interval"; in the future, the role of co-therapies with antioxidant compounds and of activation of endogeneous protective pathways such as the heme oxygenase 1 (HO-1) will need to be explored. Nitrates 31-38 heme oxygenase 1 Homo sapiens 277-293 19833168-10 2010 These results suggest that rottlerin induces up-regulation of HO-1 via PKC delta-independent pathway. rottlerin 27-36 heme oxygenase 1 Homo sapiens 62-66 19833168-11 2010 Taken together, the present study identified rottlerin as a novel inducer of HO-1 expression and identified the mechanisms involved in this process. rottlerin 45-54 heme oxygenase 1 Homo sapiens 77-81 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. rottlerin 0-9 heme oxygenase 1 Homo sapiens 36-40 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Reactive Oxygen Species 64-87 heme oxygenase 1 Homo sapiens 18-34 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Reactive Oxygen Species 64-87 heme oxygenase 1 Homo sapiens 36-40 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Reactive Oxygen Species 89-92 heme oxygenase 1 Homo sapiens 18-34 19833168-0 2010 Rottlerin induces heme oxygenase-1 (HO-1) up-regulation through reactive oxygen species (ROS) dependent and PKC delta-independent pathway in human colon cancer HT29 cells. Reactive Oxygen Species 89-92 heme oxygenase 1 Homo sapiens 36-40 19833168-1 2010 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by its substrate heme and diverse stimuli. Heme 78-82 heme oxygenase 1 Homo sapiens 0-16 19833168-1 2010 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by its substrate heme and diverse stimuli. Heme 78-82 heme oxygenase 1 Homo sapiens 18-22 19833168-3 2010 In this study, the effect of rottlerin, a putative PKC delta inhibitor, on HO-1 expression in HT29 human colon cancer cells was investigated. rottlerin 29-38 heme oxygenase 1 Homo sapiens 75-79 19833168-4 2010 Rottlerin-induced HO-1 at both protein and mRNA levels in a dose- and time-dependent manner. rottlerin 0-9 heme oxygenase 1 Homo sapiens 18-22 19833168-5 2010 Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). rottlerin 0-9 heme oxygenase 1 Homo sapiens 19-23 19833168-5 2010 Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). Acetylcysteine 67-83 heme oxygenase 1 Homo sapiens 19-23 19833168-5 2010 Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). Acetylcysteine 85-88 heme oxygenase 1 Homo sapiens 19-23 19833168-5 2010 Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). Glutathione 93-104 heme oxygenase 1 Homo sapiens 19-23 19833168-5 2010 Rottlerin-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine (NAC) or glutathione (GSH). Glutathione 106-109 heme oxygenase 1 Homo sapiens 19-23 19833168-8 2010 The pharmacological inhibition of ERK and p38 MAPK inhibited rottlerin-induced HO-1 up-regulation. rottlerin 61-70 heme oxygenase 1 Homo sapiens 79-83 21048309-0 2010 Geniposide induces the expression of heme oxygenase-1 via PI3K/Nrf2-signaling to enhance the antioxidant capacity in primary hippocampal neurons. geniposide 0-10 heme oxygenase 1 Homo sapiens 37-53 21048309-5 2010 Taken together, the novel cytoprotective mechanism of geniposide to antagonize oxidative stress may be involved in PI3K- and Nrf2-mediated upregulation of the antioxidative enzyme HO-1. geniposide 54-64 heme oxygenase 1 Homo sapiens 180-184 21048309-3 2010 In this study, geniposide upregulates the expression of heme oxygenase-1 (HO-1) to attenuate the cell apoptosis induced by 3-morpholinosydnonimine hydrochloride (SIN-1) in primary cultured hippocampal neurons. geniposide 15-25 heme oxygenase 1 Homo sapiens 56-72 21048309-3 2010 In this study, geniposide upregulates the expression of heme oxygenase-1 (HO-1) to attenuate the cell apoptosis induced by 3-morpholinosydnonimine hydrochloride (SIN-1) in primary cultured hippocampal neurons. geniposide 15-25 heme oxygenase 1 Homo sapiens 74-78 21048309-3 2010 In this study, geniposide upregulates the expression of heme oxygenase-1 (HO-1) to attenuate the cell apoptosis induced by 3-morpholinosydnonimine hydrochloride (SIN-1) in primary cultured hippocampal neurons. linsidomine 123-160 heme oxygenase 1 Homo sapiens 56-72 21048309-3 2010 In this study, geniposide upregulates the expression of heme oxygenase-1 (HO-1) to attenuate the cell apoptosis induced by 3-morpholinosydnonimine hydrochloride (SIN-1) in primary cultured hippocampal neurons. linsidomine 123-160 heme oxygenase 1 Homo sapiens 74-78 21048309-4 2010 Furthermore, geniposide induces the nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) and activation of phosphatidylinositol 3"-kinase (PI3K) in the presence of oxidative stress, and both LY294002 (a specific inhibitor of PI3K) and Zinc protoporphyrin (ZnPP, an inhibitor of HO-1) decrease the cytoprotective action of geniposide in hippocampal neurons. geniposide 13-23 heme oxygenase 1 Homo sapiens 292-296 20054154-4 2010 CdCl(2) induced effects following either apical or basolateral exposure were evaluated by Neutral Red Uptake (NRU), Trans-Epithelial Electrical Resistance (TEER), and alteration in Metallothionein 1X (MT1X), Heat shock protein 70 (HSP70), and Heme oxygenase 1 (HMOX-1) genes. cdcl 0-4 heme oxygenase 1 Homo sapiens 243-259 20378994-0 2010 Oligosaccharides from agar inhibit pro-inflammatory mediator release by inducing heme oxygenase 1. Oligosaccharides 0-16 heme oxygenase 1 Homo sapiens 81-97 20378994-0 2010 Oligosaccharides from agar inhibit pro-inflammatory mediator release by inducing heme oxygenase 1. Agar 22-26 heme oxygenase 1 Homo sapiens 81-97 20378994-3 2010 We also demonstrate that those effects of agaro-oligosaccharides on activated monocytes and macrophages may have been caused by heme oxygenase-1 induction. agaro-oligosaccharides 42-64 heme oxygenase 1 Homo sapiens 128-144 20054154-7 2010 Co-administration of N-Acetylcysteine (NAC) exerted a strong protective effect against CdCl(2) induced barrier damage and stress related genes, while other antioxidants only attenuated CdCl(2) induced HSP70 and HMOX-1 and showed no protective effect on the barrier collapse. Acetylcysteine 39-42 heme oxygenase 1 Homo sapiens 211-217 20054154-9 2010 The protective effect of NAC against CdCl(2) induced MT1X, HSP70 and HMOX-1 genes, demonstrates an anti-oxidant effect of NAC in addition to Cd chelation. Acetylcysteine 25-28 heme oxygenase 1 Homo sapiens 69-75 20054154-9 2010 The protective effect of NAC against CdCl(2) induced MT1X, HSP70 and HMOX-1 genes, demonstrates an anti-oxidant effect of NAC in addition to Cd chelation. cdcl 37-41 heme oxygenase 1 Homo sapiens 69-75 20054154-9 2010 The protective effect of NAC against CdCl(2) induced MT1X, HSP70 and HMOX-1 genes, demonstrates an anti-oxidant effect of NAC in addition to Cd chelation. Acetylcysteine 122-125 heme oxygenase 1 Homo sapiens 69-75 20054154-9 2010 The protective effect of NAC against CdCl(2) induced MT1X, HSP70 and HMOX-1 genes, demonstrates an anti-oxidant effect of NAC in addition to Cd chelation. Cadmium 37-39 heme oxygenase 1 Homo sapiens 69-75 20054154-4 2010 CdCl(2) induced effects following either apical or basolateral exposure were evaluated by Neutral Red Uptake (NRU), Trans-Epithelial Electrical Resistance (TEER), and alteration in Metallothionein 1X (MT1X), Heat shock protein 70 (HSP70), and Heme oxygenase 1 (HMOX-1) genes. cdcl 0-4 heme oxygenase 1 Homo sapiens 261-267 20054154-5 2010 CdCl(2) exposure resulted in a collapse of barrier function and the induction of MT1X, HMOX-1 and HSP70 genes, prior to alterations in cell viability. Cadmium Chloride 0-7 heme oxygenase 1 Homo sapiens 87-93 20054154-7 2010 Co-administration of N-Acetylcysteine (NAC) exerted a strong protective effect against CdCl(2) induced barrier damage and stress related genes, while other antioxidants only attenuated CdCl(2) induced HSP70 and HMOX-1 and showed no protective effect on the barrier collapse. Acetylcysteine 21-37 heme oxygenase 1 Homo sapiens 211-217 20184791-3 2010 Compared to HO-2, which is constitutively expressed, HO-1 is a stressresponsive protein that is highly induced by many agents, including cytokines, endotoxin, heavy metals, nitric oxide and its own substrate heme. Nitric Oxide 173-185 heme oxygenase 1 Homo sapiens 53-57 20064636-0 2010 Induction of heme oxygenase-1 in normal and malignant B lymphocytes by 15-deoxy-Delta(12,14)-prostaglandin J(2) requires Nrf2. 15-deoxy-delta(12,14)-prostaglandin j 71-108 heme oxygenase 1 Homo sapiens 13-29 20064636-5 2010 15d-PGJ(2) potently induced HO-1 protein expression in normal and malignant B cells. 15d-pgj 0-7 heme oxygenase 1 Homo sapiens 28-32 20184791-3 2010 Compared to HO-2, which is constitutively expressed, HO-1 is a stressresponsive protein that is highly induced by many agents, including cytokines, endotoxin, heavy metals, nitric oxide and its own substrate heme. Heme 208-212 heme oxygenase 1 Homo sapiens 53-57 19954435-1 2010 X-ray crystal structure of human heme oxygenase-1 in complex with 4-phenyl-1-(1H-1,2,4-triazol-1-yl)-2-butanone. 4-phenyl-1-(1H-1,2,4-triazol-1-yl)-2-butanone 66-111 heme oxygenase 1 Homo sapiens 33-49 20184791-5 2010 Over the past decade, compelling evidence has revealed that the induction of HO-1 represents an important defensive mechanism against further oxidative injury in tissues and cells following various insults; this occurs by virtue of the anti-inflammatory and antioxidant capacities of CO, biliverdin, and the subsequent metabolite of biliverdin, bilirubin. Carbon Monoxide 284-286 heme oxygenase 1 Homo sapiens 77-81 19954435-2 2010 A series of 1-azolyl-4-phenyl-2-butanones was designed and synthesized for the inhibition of heme oxygenases (heme oxygenase-1 and heme oxygenase-2). 1-azolyl-4-phenyl-2-butanones 12-41 heme oxygenase 1 Homo sapiens 110-147 20184791-5 2010 Over the past decade, compelling evidence has revealed that the induction of HO-1 represents an important defensive mechanism against further oxidative injury in tissues and cells following various insults; this occurs by virtue of the anti-inflammatory and antioxidant capacities of CO, biliverdin, and the subsequent metabolite of biliverdin, bilirubin. Biliverdine 288-298 heme oxygenase 1 Homo sapiens 77-81 19954435-7 2010 The first reported X-ray crystal structure of human heme oxygenase-1 in complex with a 1,2,4-triazole-based inhibitor, namely 4-phenyl-1-(1H-1,2,4-triazol-1-yl)-2-butanone, was also determined. 1,2,4-triazole 87-101 heme oxygenase 1 Homo sapiens 52-68 20184791-5 2010 Over the past decade, compelling evidence has revealed that the induction of HO-1 represents an important defensive mechanism against further oxidative injury in tissues and cells following various insults; this occurs by virtue of the anti-inflammatory and antioxidant capacities of CO, biliverdin, and the subsequent metabolite of biliverdin, bilirubin. Biliverdine 333-343 heme oxygenase 1 Homo sapiens 77-81 19954435-7 2010 The first reported X-ray crystal structure of human heme oxygenase-1 in complex with a 1,2,4-triazole-based inhibitor, namely 4-phenyl-1-(1H-1,2,4-triazol-1-yl)-2-butanone, was also determined. 4-phenyl-1-(1H-1,2,4-triazol-1-yl)-2-butanone 126-171 heme oxygenase 1 Homo sapiens 52-68 20184791-5 2010 Over the past decade, compelling evidence has revealed that the induction of HO-1 represents an important defensive mechanism against further oxidative injury in tissues and cells following various insults; this occurs by virtue of the anti-inflammatory and antioxidant capacities of CO, biliverdin, and the subsequent metabolite of biliverdin, bilirubin. Bilirubin 345-354 heme oxygenase 1 Homo sapiens 77-81 19954435-8 2010 The inhibitor binds to the human heme oxygenase-1 distal pocket through the coordination of heme iron by the N4 in the triazole moiety, whereas the phenyl group is stabilized by hydrophobic interactions from residues within the binding pocket. Iron 97-101 heme oxygenase 1 Homo sapiens 33-49 19954435-8 2010 The inhibitor binds to the human heme oxygenase-1 distal pocket through the coordination of heme iron by the N4 in the triazole moiety, whereas the phenyl group is stabilized by hydrophobic interactions from residues within the binding pocket. Folic Acid 109-111 heme oxygenase 1 Homo sapiens 33-49 21525764-9 2010 RESULTS: Ethanol and TGF-beta rapidly increase ROI and reduce GSH in hHeps, causing apoptosis with a release of approximately 40% total LDH after 72 h. Similar to incubation with hemin preincubation and co-incubation of cells with nifedipine, verapamil and quercetin significantly reduce oxidative stress and resulting cellular damage, in a dose-dependent manner, by initiating nuclear translocation of Nrf2 which in turn induces HO-1 under the control of p38 and ERK. Ethanol 9-16 heme oxygenase 1 Homo sapiens 430-434 19954435-8 2010 The inhibitor binds to the human heme oxygenase-1 distal pocket through the coordination of heme iron by the N4 in the triazole moiety, whereas the phenyl group is stabilized by hydrophobic interactions from residues within the binding pocket. Triazoles 119-127 heme oxygenase 1 Homo sapiens 33-49 21525764-10 2010 Blocking of HO-1 activity with ZNPP9 reverses the protective effect of all three substances. zinc protoporphyrin 31-36 heme oxygenase 1 Homo sapiens 12-16 21525764-12 2010 As polyphenols have great potential to induce HO-1 expression, they may play an important role for future therapeutic strategies to protect liver from oxidative stress-dependent damage observed during chronic alcohol consumption. Polyphenols 3-14 heme oxygenase 1 Homo sapiens 46-50 21525764-12 2010 As polyphenols have great potential to induce HO-1 expression, they may play an important role for future therapeutic strategies to protect liver from oxidative stress-dependent damage observed during chronic alcohol consumption. Alcohols 209-216 heme oxygenase 1 Homo sapiens 46-50 19720622-3 2010 We have recently shown that UVA-1 oxidized the abundant membrane phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC), which then induced the stress-response protein heme oxygenase 1 (HO-1) in dermal fibroblasts. phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine 65-135 heme oxygenase 1 Homo sapiens 191-207 19720622-3 2010 We have recently shown that UVA-1 oxidized the abundant membrane phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC), which then induced the stress-response protein heme oxygenase 1 (HO-1) in dermal fibroblasts. phospholipid 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine 65-135 heme oxygenase 1 Homo sapiens 209-213 20700711-3 2010 Quantitative PCR analysis as well as Western blotting demonstrated a specific upregulation of Hsp70 mRNA and protein after heat shock, while heme oxygenase 1 (HO1 or Hsp32) mRNA and protein was specifically upregulated following H2O2-induced oxidative stress. Hydrogen Peroxide 229-233 heme oxygenase 1 Homo sapiens 141-157 20388101-4 2010 The mechanisms through which dietary supplementation with antioxidants may be useful to prevent free radical-related diseases is related to their ability to counteract toxic production of both reactive oxygen and nitrogen species, along with the up-regulation of vitagenes, such as members of the heat shock protein (Hsp) family heme oxygenase-1 and Hsp70. Free Radicals 96-108 heme oxygenase 1 Homo sapiens 329-345 19375813-3 2010 We found that treatment with cisplatin further augmented HO-1 expression, which was associated with activation of the epidermal growth factor receptor (EGFR) mediated signaling pathway and subsequent nuclear translocation of NF-kappaB. Cisplatin 29-38 heme oxygenase 1 Homo sapiens 57-61 19375813-4 2010 In concordance with the findings, treatment with EGFR-selective tyrosine kinase inhibitor (AG1478) or an Akt inhibitor, which interfere with the post-EGFR signaling pathway, suppressed cisplatin induced HO-1 expression. RTKI cpd 91-97 heme oxygenase 1 Homo sapiens 203-207 19375813-4 2010 In concordance with the findings, treatment with EGFR-selective tyrosine kinase inhibitor (AG1478) or an Akt inhibitor, which interfere with the post-EGFR signaling pathway, suppressed cisplatin induced HO-1 expression. Cisplatin 185-194 heme oxygenase 1 Homo sapiens 203-207 19375813-7 2010 Collectively, the results indicate that resistance to cisplatin in A549 cells is associated with HO-1 through EGFR mediated signaling pathway including activation of the PI3k/Akt and NF-kappaB systems. Cisplatin 54-63 heme oxygenase 1 Homo sapiens 97-101 21504135-11 2010 Consequently, the expression of heme oxygenase-1 and gamma-glutamyl-cysteine-synthase were elevated after 24 hrs of acrolein treatment. Acrolein 116-124 heme oxygenase 1 Homo sapiens 32-48 19375813-2 2010 We investigated involvement of HO-1 in chemoresistance of cisplatin in human lung epithelial adenocarcinoma cell line, A549, which constitutively expressed HO-1. Cisplatin 58-67 heme oxygenase 1 Homo sapiens 31-35 20072927-0 2010 Characterization of the antioxidant properties of pentaerithrityl tetranitrate (PETN)-induction of the intrinsic antioxidative system heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 50-78 heme oxygenase 1 Homo sapiens 134-150 20700711-3 2010 Quantitative PCR analysis as well as Western blotting demonstrated a specific upregulation of Hsp70 mRNA and protein after heat shock, while heme oxygenase 1 (HO1 or Hsp32) mRNA and protein was specifically upregulated following H2O2-induced oxidative stress. Hydrogen Peroxide 229-233 heme oxygenase 1 Homo sapiens 159-162 20072927-0 2010 Characterization of the antioxidant properties of pentaerithrityl tetranitrate (PETN)-induction of the intrinsic antioxidative system heme oxygenase-1 (HO-1). Pentaerythritol Tetranitrate 80-84 heme oxygenase 1 Homo sapiens 134-150 20700711-3 2010 Quantitative PCR analysis as well as Western blotting demonstrated a specific upregulation of Hsp70 mRNA and protein after heat shock, while heme oxygenase 1 (HO1 or Hsp32) mRNA and protein was specifically upregulated following H2O2-induced oxidative stress. Hydrogen Peroxide 229-233 heme oxygenase 1 Homo sapiens 166-171 20072927-6 2010 Some of these effects are related to special pharmacokinetics of PETN, but upon chronic administration, PETN also induces antioxidative pathways at the genomic level, resulting in increased expression of heme oxygenase-1 (HO-1) and ferritin, both possessing highly protective properties. Pentaerythritol Tetranitrate 104-108 heme oxygenase 1 Homo sapiens 204-220 19591088-0 2010 Protective effect of sauchinone by upregulating heme oxygenase-1 via the P38 MAPK and Nrf2/ARE pathways in HepG2 cells. sauchinone 21-31 heme oxygenase 1 Homo sapiens 48-64 19895178-2 2010 In this study, the correlation between the HO-1 gene promoter polymorphism and alcohol, along with the risk of ESCC on Chinese males, was analyzed.The case-control study was performed in 143 ESCC patients and 264 cancer-free controls. Alcohols 79-86 heme oxygenase 1 Homo sapiens 43-47 20062840-4 2010 Furthermore, a reduction of free heme levels led to a strong decrease in UVA-induced Nrf2 and HO-1 protein levels confirming a clear role for heme in the UV-mediated stress response. Heme 33-37 heme oxygenase 1 Homo sapiens 94-98 20062840-4 2010 Furthermore, a reduction of free heme levels led to a strong decrease in UVA-induced Nrf2 and HO-1 protein levels confirming a clear role for heme in the UV-mediated stress response. Heme 142-146 heme oxygenase 1 Homo sapiens 94-98 19591088-4 2010 This study investigated the potency of sauchinone as a hepatic heme oxygenase (HO)-1 inducer and its protective effects in HepG2 cells. sauchinone 39-49 heme oxygenase 1 Homo sapiens 63-84 20199188-5 2010 Interestingly, inhibition of HO-1 induction (by zinc protoporphyrin, ZnP) was associated with an accelerated mitogenic response to EGF and HGF in MCs. zinc protoporphyrin 48-67 heme oxygenase 1 Homo sapiens 29-33 21062534-4 2010 In fact, it has been demonstrated that the activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in various animal intestinal injury models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid or dextran sulfate sodium. Indomethacin 224-236 heme oxygenase 1 Homo sapiens 57-61 21062534-4 2010 In fact, it has been demonstrated that the activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in various animal intestinal injury models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid or dextran sulfate sodium. Trinitrobenzenesulfonic Acid 276-305 heme oxygenase 1 Homo sapiens 57-61 21062534-4 2010 In fact, it has been demonstrated that the activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in various animal intestinal injury models induced by ischemia-reperfusion, indomethacin, lipopolysaccharide-associated sepsis, trinitrobenzene sulfonic acid or dextran sulfate sodium. Dextran Sulfate 309-331 heme oxygenase 1 Homo sapiens 57-61 21062534-5 2010 In addition, carbon monoxide (CO) derived from HO-1 has been shown to be involved in the regulation of intestinal inflammation. Carbon Monoxide 13-28 heme oxygenase 1 Homo sapiens 47-51 21062534-5 2010 In addition, carbon monoxide (CO) derived from HO-1 has been shown to be involved in the regulation of intestinal inflammation. Carbon Monoxide 30-32 heme oxygenase 1 Homo sapiens 47-51 20199188-5 2010 Interestingly, inhibition of HO-1 induction (by zinc protoporphyrin, ZnP) was associated with an accelerated mitogenic response to EGF and HGF in MCs. pyrithione zinc 69-72 heme oxygenase 1 Homo sapiens 29-33 19781563-0 2010 Ginsenoside Rb1 protects against 6-hydroxydopamine-induced oxidative stress by increasing heme oxygenase-1 expression through an estrogen receptor-related PI3K/Akt/Nrf2-dependent pathway in human dopaminergic cells. Oxidopamine 33-50 heme oxygenase 1 Homo sapiens 90-106 19781563-5 2010 In this study, we demonstrate that the phytoestrogen Rb1 inhibits 6-hydroxydopamine (6-OHDA)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. Oxidopamine 66-83 heme oxygenase 1 Homo sapiens 158-174 20172305-2 2010 The mechanisms responsible for this effective graft protection against ischemia-reperfusion injury are pivotal actions on generation of nitric oxide a diffusible molecule with vasodilator properties, that facilitates the up-regulation of other well-known cytoprotective genes, such as hypoxia-inducible factor-1 alpha (HIF-1alpha) and heme-oxygenase 1 (HO-1). Nitric Oxide 136-148 heme oxygenase 1 Homo sapiens 335-351 20172305-2 2010 The mechanisms responsible for this effective graft protection against ischemia-reperfusion injury are pivotal actions on generation of nitric oxide a diffusible molecule with vasodilator properties, that facilitates the up-regulation of other well-known cytoprotective genes, such as hypoxia-inducible factor-1 alpha (HIF-1alpha) and heme-oxygenase 1 (HO-1). Nitric Oxide 136-148 heme oxygenase 1 Homo sapiens 353-357 19781563-5 2010 In this study, we demonstrate that the phytoestrogen Rb1 inhibits 6-hydroxydopamine (6-OHDA)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. Oxidopamine 85-91 heme oxygenase 1 Homo sapiens 158-174 20172305-3 2010 During normoxic reperfusion, the presence of nitric oxide permits HIF-1alpha accumulation to inhibit prolyl-hydoxylases, thus promoting an additional overexpression of the HO-1 in steatotic and nonsteatotic graft livers preserved in IGL-1. Nitric Oxide 45-57 heme oxygenase 1 Homo sapiens 172-176 20175232-7 2010 CONCLUSION: DRB and alpha-Amanitin can down-regulate the expression of Nrf2 and its targeting proteins HO-1, AKR1C and NQO1, but may have no effect on the localization of Nrf2. Alpha-Amanitin 20-34 heme oxygenase 1 Homo sapiens 103-114 20193368-1 2009 OBJECTIVE: To investigate the effects of alpha-zearalanol on the expression of heme oxygenase-1 (HO-1) gene and cytosolic free calcium level in the tumor necrosis factor alpha (TNF-alpha)-stimulated human umbilical vein endothelial cell (HUVEC) and the mechanisms involved. Zeranol 41-57 heme oxygenase 1 Homo sapiens 79-95 20193368-1 2009 OBJECTIVE: To investigate the effects of alpha-zearalanol on the expression of heme oxygenase-1 (HO-1) gene and cytosolic free calcium level in the tumor necrosis factor alpha (TNF-alpha)-stimulated human umbilical vein endothelial cell (HUVEC) and the mechanisms involved. Zeranol 41-57 heme oxygenase 1 Homo sapiens 97-101 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. Zeranol 169-185 heme oxygenase 1 Homo sapiens 29-33 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. Zeranol 169-185 heme oxygenase 1 Homo sapiens 144-148 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. Zeranol 169-185 heme oxygenase 1 Homo sapiens 144-148 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. Zeranol 169-185 heme oxygenase 1 Homo sapiens 144-148 20733275-6 2010 In our analysis of 16 cases treated by intravesical administration of anthracyclines, the positive expression of HO-1 correlated with poor disease-free survival (p = 0.015). Anthracyclines 70-84 heme oxygenase 1 Homo sapiens 113-117 20733275-7 2010 In in vitro experiments using urothelial cancer cell lines, HO-1 upregulation was observed by exposure to doxorubicin. Doxorubicin 106-117 heme oxygenase 1 Homo sapiens 60-64 20733275-8 2010 Moreover, siRNA-mediated suppression of HO-1 upregulation sensitized the urothelial cancer cells to doxorubicin. Doxorubicin 100-111 heme oxygenase 1 Homo sapiens 40-44 20733275-9 2010 CONCLUSIONS: Our findings suggested that resistance against anthracyclines correlated with HO-1 and expression analysis of HO-1 may be a useful predictive marker for intravesical administration of anthracyclines. Anthracyclines 60-74 heme oxygenase 1 Homo sapiens 91-95 20733275-9 2010 CONCLUSIONS: Our findings suggested that resistance against anthracyclines correlated with HO-1 and expression analysis of HO-1 may be a useful predictive marker for intravesical administration of anthracyclines. Anthracyclines 197-211 heme oxygenase 1 Homo sapiens 123-127 20193368-0 2009 [Effect of alpha-zearalanol upon the expression of HO-1 gene and the cytosolic free calcium level in tumor necrosis factor alpha-stimulated human endothelial cell]. Zeranol 11-27 heme oxygenase 1 Homo sapiens 51-55 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. alpha-zal 270-279 heme oxygenase 1 Homo sapiens 29-33 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. alpha-zal 270-279 heme oxygenase 1 Homo sapiens 144-148 19952508-2 2009 CdCl(2), heme, and diclofenac sodium salt (diclofenac) were used as inducers of HO-1. Cadmium Chloride 0-7 heme oxygenase 1 Homo sapiens 80-84 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. alpha-zal 270-279 heme oxygenase 1 Homo sapiens 144-148 20193368-8 2009 TNF-alpha stimulation caused HO-1 mRNA increased by 145% of control (0.88 +/- 0.10 vs 0.36 +/- 0.11, P < 0.01), and also obviously increased HO-1 protein expression; alpha-zearalanol inhibited the expression of HO-1 mRNA in a dose-dependent manner; Pretreatment with alpha-ZAL and p47(phox) siRNA both attenuated TNFalpha-induced HO-1 protein expression. alpha-zal 270-279 heme oxygenase 1 Homo sapiens 144-148 20193368-10 2009 CONCLUSION: ROS only partly mediated HO-1; expression in the TNF-alpha-stimulated HUVEC; alpha-ZAL has a potent inhibitory effect on the HO-1 expression and cytosolic free calcium level in the TNF-alpha-stimulated HUVEC, mainly through the inhibition of ROS generation derived from NADPH oxidase. ros 12-15 heme oxygenase 1 Homo sapiens 37-41 20193368-10 2009 CONCLUSION: ROS only partly mediated HO-1; expression in the TNF-alpha-stimulated HUVEC; alpha-ZAL has a potent inhibitory effect on the HO-1 expression and cytosolic free calcium level in the TNF-alpha-stimulated HUVEC, mainly through the inhibition of ROS generation derived from NADPH oxidase. ros 12-15 heme oxygenase 1 Homo sapiens 137-141 20193368-10 2009 CONCLUSION: ROS only partly mediated HO-1; expression in the TNF-alpha-stimulated HUVEC; alpha-ZAL has a potent inhibitory effect on the HO-1 expression and cytosolic free calcium level in the TNF-alpha-stimulated HUVEC, mainly through the inhibition of ROS generation derived from NADPH oxidase. alpha-zal 89-98 heme oxygenase 1 Homo sapiens 37-41 20193368-10 2009 CONCLUSION: ROS only partly mediated HO-1; expression in the TNF-alpha-stimulated HUVEC; alpha-ZAL has a potent inhibitory effect on the HO-1 expression and cytosolic free calcium level in the TNF-alpha-stimulated HUVEC, mainly through the inhibition of ROS generation derived from NADPH oxidase. alpha-zal 89-98 heme oxygenase 1 Homo sapiens 137-141 19909254-5 2009 HO-1 (haem oxygenase-1), an anti-inflammatory enzyme, and its metabolite, CO (carbon monoxide), exert protective effects in several organs against oxidative stimuli. Carbon Monoxide 74-76 heme oxygenase 1 Homo sapiens 0-4 19909254-5 2009 HO-1 (haem oxygenase-1), an anti-inflammatory enzyme, and its metabolite, CO (carbon monoxide), exert protective effects in several organs against oxidative stimuli. Carbon Monoxide 74-76 heme oxygenase 1 Homo sapiens 6-22 19909254-5 2009 HO-1 (haem oxygenase-1), an anti-inflammatory enzyme, and its metabolite, CO (carbon monoxide), exert protective effects in several organs against oxidative stimuli. Carbon Monoxide 78-93 heme oxygenase 1 Homo sapiens 0-4 19909254-5 2009 HO-1 (haem oxygenase-1), an anti-inflammatory enzyme, and its metabolite, CO (carbon monoxide), exert protective effects in several organs against oxidative stimuli. Carbon Monoxide 78-93 heme oxygenase 1 Homo sapiens 6-22 19735655-4 2009 Chemical inhibition of HO-1 with zinc protoporphyrin-IX prevents neuroprotection by MGF24 against the three neurotoxins. zinc protoporphyrin 33-55 heme oxygenase 1 Homo sapiens 23-27 19961388-0 2009 The anti-inflammatory role of heme oxygenase-1 in lipopolysaccharide and cytokine-stimulated inducible nitric oxide synthase and nitric oxide production in human periodontal ligament cells. Nitric Oxide 103-115 heme oxygenase 1 Homo sapiens 30-46 19961388-2 2009 Therefore, the aim of this study is to investigate the anti-inflammatory properties of HO-1 in lipopolysaccharide (LPS)- and proinflammatory cytokine-stimulated inducible nitric oxide synthase (iNOS) expression and nitric oxide (NO) production in human periodontal ligament (PDL) cells. Nitric Oxide 171-183 heme oxygenase 1 Homo sapiens 87-91 20608094-4 2009 Expressions of biopyrrins and heme oxygenase-1 (HO-1), a stress-responsive bilirubin-producing enzyme, in heart, aorta, kidney, liver and lung were immunostained with autopsied specimens. Bilirubin 75-84 heme oxygenase 1 Homo sapiens 30-46 20608094-4 2009 Expressions of biopyrrins and heme oxygenase-1 (HO-1), a stress-responsive bilirubin-producing enzyme, in heart, aorta, kidney, liver and lung were immunostained with autopsied specimens. Bilirubin 75-84 heme oxygenase 1 Homo sapiens 48-52 20608094-12 2009 Induction of anti-oxidative enzyme HO-1 seemed to be involved in the activation of bilirubin/biopyrrin pathway. Bilirubin 83-92 heme oxygenase 1 Homo sapiens 35-39 20608094-12 2009 Induction of anti-oxidative enzyme HO-1 seemed to be involved in the activation of bilirubin/biopyrrin pathway. biopyrrin 93-102 heme oxygenase 1 Homo sapiens 35-39 19952508-2 2009 CdCl(2), heme, and diclofenac sodium salt (diclofenac) were used as inducers of HO-1. Diclofenac 19-41 heme oxygenase 1 Homo sapiens 80-84 19952508-2 2009 CdCl(2), heme, and diclofenac sodium salt (diclofenac) were used as inducers of HO-1. Diclofenac 19-29 heme oxygenase 1 Homo sapiens 80-84 19952508-6 2009 HO-1 levels were increased by CdCl(2) (7.5 microM), heme (10, 100 microM), and stannic mesoporphyrin (SnMP) (10 microM), but were not changed by AAP, and were decreased by diclofenac. cdcl 30-34 heme oxygenase 1 Homo sapiens 0-4 19952508-6 2009 HO-1 levels were increased by CdCl(2) (7.5 microM), heme (10, 100 microM), and stannic mesoporphyrin (SnMP) (10 microM), but were not changed by AAP, and were decreased by diclofenac. Heme 52-56 heme oxygenase 1 Homo sapiens 0-4 19952508-6 2009 HO-1 levels were increased by CdCl(2) (7.5 microM), heme (10, 100 microM), and stannic mesoporphyrin (SnMP) (10 microM), but were not changed by AAP, and were decreased by diclofenac. mesoporphyrin IX 87-100 heme oxygenase 1 Homo sapiens 0-4 19952508-6 2009 HO-1 levels were increased by CdCl(2) (7.5 microM), heme (10, 100 microM), and stannic mesoporphyrin (SnMP) (10 microM), but were not changed by AAP, and were decreased by diclofenac. tin mesoporphyrin 102-106 heme oxygenase 1 Homo sapiens 0-4 19952508-6 2009 HO-1 levels were increased by CdCl(2) (7.5 microM), heme (10, 100 microM), and stannic mesoporphyrin (SnMP) (10 microM), but were not changed by AAP, and were decreased by diclofenac. Diclofenac 172-182 heme oxygenase 1 Homo sapiens 0-4 19952508-7 2009 HO-1 levels were increased by diclofenac (300 microM), and CP (36 microM), but were unaffected by AAP because of low sensitivity in HepG2 cells. Diclofenac 30-40 heme oxygenase 1 Homo sapiens 0-4 19734319-7 2009 Upregulation of HO-1 is not inhibited by Trolox, a non-thiol antioxidant, and does not involve the transcription factors AP-1 or Nrf2. Sulfhydryl Compounds 55-60 heme oxygenase 1 Homo sapiens 16-20 19797172-10 2009 Transfection of HAECs with mTOR (mammalian target of rapamycin) short hairpin RNA and with Akt2 small interfering RNA significantly inhibited RSG-mediated transcriptional upregulation of heme oxygenase-1, a PPARgamma target gene. Rosiglitazone 142-145 heme oxygenase 1 Homo sapiens 187-203 19822148-3 2009 CS exposure led to increased HO-1 and nuclear Nrf2 expression (6 h) followed by decreased HO-1 expression concomitantly with nuclear Nrf2/Bach1 ratio decrease (72h). Cesium 0-2 heme oxygenase 1 Homo sapiens 29-33 19822148-3 2009 CS exposure led to increased HO-1 and nuclear Nrf2 expression (6 h) followed by decreased HO-1 expression concomitantly with nuclear Nrf2/Bach1 ratio decrease (72h). Cesium 0-2 heme oxygenase 1 Homo sapiens 90-94 19822148-5 2009 Extracellular-signal-regulated kinase(1/2) (ERK(1/2)) and c-Jun NH2-terminal kinase (JNK) inhibition completely abrogated CS effects on HO-1 expression and nuclear Nrf2/Bach1 translocation. Cesium 122-124 heme oxygenase 1 Homo sapiens 136-140 19822148-6 2009 These results suggest that ERK(1/2) and JNK are involved in CS-induced biphasic HO-1 expression by a specific regulation of Nrf2/Keap1-Bach1. Cesium 60-62 heme oxygenase 1 Homo sapiens 80-84 19615435-3 2009 Comparison of the in vitro neurotoxicity data with general cytotoxicity data generated in a non-neuronal cell line and with in vivo data such as acute human lethal blood concentration, revealed that GABA(A) receptor function, acetylcholine esterase activity, cell membrane potential, glucose uptake, total RNA expression and altered gene expression of NF-H, GFAP, MBP, HSP32 and caspase-3 were the best endpoints to use for further testing with 36 additional chemicals. gamma-Aminobutyric Acid 199-203 heme oxygenase 1 Homo sapiens 369-374 19797172-11 2009 Chromatin immunoprecipitation assay demonstrated sirolimus interferes with binding of PPARgamma to its response elements in heme oxygenase-1 promoter. Sirolimus 49-58 heme oxygenase 1 Homo sapiens 124-140 19734319-0 2009 Peroxisome proliferator-activated receptor-gamma ligands induce heme oxygenase-1 in lung fibroblasts by a PPARgamma-independent, glutathione-dependent mechanism. Glutathione 129-140 heme oxygenase 1 Homo sapiens 64-80 19734319-4 2009 We report here that the PPARgamma ligands 15d-PGJ2 and 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO), which have potent antifibrotic effects in vitro, also strongly induce HO-1 expression in primary human lung fibroblasts. 15-deoxyprostaglandin J2 42-50 heme oxygenase 1 Homo sapiens 178-182 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. 15-deoxyprostaglandin J2 51-59 heme oxygenase 1 Homo sapiens 76-80 19734319-4 2009 We report here that the PPARgamma ligands 15d-PGJ2 and 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO), which have potent antifibrotic effects in vitro, also strongly induce HO-1 expression in primary human lung fibroblasts. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 55-99 heme oxygenase 1 Homo sapiens 178-182 19734319-4 2009 We report here that the PPARgamma ligands 15d-PGJ2 and 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO), which have potent antifibrotic effects in vitro, also strongly induce HO-1 expression in primary human lung fibroblasts. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 101-105 heme oxygenase 1 Homo sapiens 178-182 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Glutathione 60-71 heme oxygenase 1 Homo sapiens 16-20 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Glutathione 73-76 heme oxygenase 1 Homo sapiens 16-20 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Acetylcysteine 109-126 heme oxygenase 1 Homo sapiens 16-20 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Acetylcysteine 128-131 heme oxygenase 1 Homo sapiens 16-20 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Sulfhydryl Compounds 136-141 heme oxygenase 1 Homo sapiens 16-20 19734319-6 2009 Upregulation of HO-1 coincides with decreased intracellular glutathione (GSH) levels and can be inhibited by N-acetyl cysteine (NAC), a thiol antioxidant and GSH precursor. Glutathione 158-161 heme oxygenase 1 Homo sapiens 16-20 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 64-68 heme oxygenase 1 Homo sapiens 76-80 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. Glutathione 87-90 heme oxygenase 1 Homo sapiens 76-80 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. 15-deoxyprostaglandin J2 161-169 heme oxygenase 1 Homo sapiens 76-80 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 173-177 heme oxygenase 1 Homo sapiens 76-80 19734319-10 2009 These data suggest that in human lung fibroblasts, 15d-PGJ2 and CDDO induce HO-1 via a GSH-dependent mechanism involving the formation of covalent bonds between 15d-PGJ2 or CDDO and GSH. Glutathione 182-185 heme oxygenase 1 Homo sapiens 76-80 19734319-11 2009 Inhibiting HO-1 upregulation with NAC has only a small effect on the antifibrotic properties of 15d-PGJ2 and CDDO in vitro. Acetylcysteine 34-37 heme oxygenase 1 Homo sapiens 11-15 19734319-11 2009 Inhibiting HO-1 upregulation with NAC has only a small effect on the antifibrotic properties of 15d-PGJ2 and CDDO in vitro. 15-deoxyprostaglandin J2 96-104 heme oxygenase 1 Homo sapiens 11-15 19734319-11 2009 Inhibiting HO-1 upregulation with NAC has only a small effect on the antifibrotic properties of 15d-PGJ2 and CDDO in vitro. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 109-113 heme oxygenase 1 Homo sapiens 11-15 19729077-3 2009 MATERIALS AND METHODS: Western blotting was used to examine the effect of apigenin (10-40 microM) on the LPS- and nicotine-induced expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and heme oxygenase-1 (HO-1), as well as the phosphorylation of mitogen-activated protein kinases (MAPKs), in hPDL cells. Nicotine 114-122 heme oxygenase 1 Homo sapiens 215-231 19397697-10 2009 Induction of HMOX-1 expression by pantothenol and pantothenic acid in dermal cells was confirmed on the protein level using immunoblots. dexpanthenol 34-45 heme oxygenase 1 Homo sapiens 13-19 19397697-10 2009 Induction of HMOX-1 expression by pantothenol and pantothenic acid in dermal cells was confirmed on the protein level using immunoblots. Pantothenic Acid 50-66 heme oxygenase 1 Homo sapiens 13-19 19729077-3 2009 MATERIALS AND METHODS: Western blotting was used to examine the effect of apigenin (10-40 microM) on the LPS- and nicotine-induced expression of cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), and heme oxygenase-1 (HO-1), as well as the phosphorylation of mitogen-activated protein kinases (MAPKs), in hPDL cells. Nicotine 114-122 heme oxygenase 1 Homo sapiens 233-237 19729077-0 2009 Anti-inflammatory effects of apigenin on nicotine- and lipopolysaccharide-stimulated human periodontal ligament cells via heme oxygenase-1. Apigenin 29-37 heme oxygenase 1 Homo sapiens 122-138 19729077-5 2009 RESULTS: Incubation of hPDL cells with apigenin decreased LPS- and nicotine-induced HO-1 protein expression and activity. Nicotine 67-75 heme oxygenase 1 Homo sapiens 84-88 19729077-7 2009 Hemin, a selective HO-1 inducer, reversed the apigenin-mediated suppression of nicotine- and LPS-induced NO, PGE2 and cytokine production. Nicotine 79-87 heme oxygenase 1 Homo sapiens 19-23 19536150-2 2009 We showed by real-time PCR, western blotting, and ELISA that HO-1 mRNA and protein expression by cultured normal human keratinocytes was upregulated during epidermal differentiation induced by a high-calcium medium. Calcium 200-207 heme oxygenase 1 Homo sapiens 61-65 19903769-3 2009 Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Heme 78-82 heme oxygenase 1 Homo sapiens 0-16 19903769-3 2009 Heme oxygenase 1 (HO-1), the inducible isoform of the rate-limiting enzyme in heme degradation, counteracts oxidative and inflammatory damage. Heme 78-82 heme oxygenase 1 Homo sapiens 18-22 19903769-7 2009 Hemin treatment increased HO-1 at both protein and mRNA levels in all cell lines and decreased cell proliferation and invasion. Hemin 0-5 heme oxygenase 1 Homo sapiens 26-30 19625608-0 2009 Hypochlorous acid-induced heme oxygenase-1 gene expression promotes human endothelial cell survival. Hypochlorous Acid 0-17 heme oxygenase 1 Homo sapiens 26-42 19679656-4 2009 Elevations in mRNA and protein levels of HIF-dependent genes heme oxygenase-1 (Ho-1) and manganese superoxide dismutase (Mnsod) following DHB pretreatment alone are also maintained in the presence of MPTP. 3,4-dihydroxybenzoate 138-141 heme oxygenase 1 Homo sapiens 61-77 19679656-4 2009 Elevations in mRNA and protein levels of HIF-dependent genes heme oxygenase-1 (Ho-1) and manganese superoxide dismutase (Mnsod) following DHB pretreatment alone are also maintained in the presence of MPTP. 3,4-dihydroxybenzoate 138-141 heme oxygenase 1 Homo sapiens 79-83 19679656-4 2009 Elevations in mRNA and protein levels of HIF-dependent genes heme oxygenase-1 (Ho-1) and manganese superoxide dismutase (Mnsod) following DHB pretreatment alone are also maintained in the presence of MPTP. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 200-204 heme oxygenase 1 Homo sapiens 61-77 19679656-4 2009 Elevations in mRNA and protein levels of HIF-dependent genes heme oxygenase-1 (Ho-1) and manganese superoxide dismutase (Mnsod) following DHB pretreatment alone are also maintained in the presence of MPTP. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine 200-204 heme oxygenase 1 Homo sapiens 79-83 19686725-0 2009 Synergistic induction of heme oxygenase-1 by nicaraven after subarachnoid hemorrhage to prevent delayed cerebral vasospasm. nicaraven 45-54 heme oxygenase 1 Homo sapiens 25-41 19686725-7 2009 Antisense HO-1 oligodeoxynucleotides abrogated this HO-1 induction and the antivasospastic effect of nicaraven. nicaraven 101-110 heme oxygenase 1 Homo sapiens 10-14 19686725-8 2009 In vitro study using Hela cells, nicaraven enhanced the human HO-1 promoter (-4.5 kbp) activity, which was pre-activated with the blood component oxyhemoglobin to mimic the ability of subarachnoid hemorrhage. nicaraven 33-42 heme oxygenase 1 Homo sapiens 62-66 19686725-9 2009 These results suggest that this enhanced HO-1 expression through a combination of pathological state and pharmacological agent could be an effective strategy to improve the prognosis of heme- and oxidative stress-induced diseases, such as delayed cerebral vasospasm. Heme 186-190 heme oxygenase 1 Homo sapiens 41-45 19625608-3 2009 Treatment of human endothelial cells with the myeloperoxidase product HOCl stimulated a concentration- and time-dependent increase in HO-1 protein that resulted in a significant rise in carbon monoxide (CO) production. Carbon Monoxide 186-201 heme oxygenase 1 Homo sapiens 134-138 19625608-3 2009 Treatment of human endothelial cells with the myeloperoxidase product HOCl stimulated a concentration- and time-dependent increase in HO-1 protein that resulted in a significant rise in carbon monoxide (CO) production. Carbon Monoxide 203-205 heme oxygenase 1 Homo sapiens 134-138 19625608-5 2009 In addition, HOCl induced a significant rise in HO-1 promoter activity that was blocked by mutating the antioxidant response element (ARE) in the promoter or by overexpressing a dominant-negative mutant of Nrf2. Hypochlorous Acid 13-17 heme oxygenase 1 Homo sapiens 48-52 19625608-6 2009 The HOCl-mediated induction of Nrf2 or HO-1 was blocked by the glutathione donor N-acetyl-l-cysteine but was unaffected by ascorbic or uric acid. Hypochlorous Acid 4-8 heme oxygenase 1 Homo sapiens 39-43 19625608-6 2009 The HOCl-mediated induction of Nrf2 or HO-1 was blocked by the glutathione donor N-acetyl-l-cysteine but was unaffected by ascorbic or uric acid. Glutathione 63-74 heme oxygenase 1 Homo sapiens 39-43 19625608-6 2009 The HOCl-mediated induction of Nrf2 or HO-1 was blocked by the glutathione donor N-acetyl-l-cysteine but was unaffected by ascorbic or uric acid. Acetylcysteine 81-100 heme oxygenase 1 Homo sapiens 39-43 19625608-7 2009 Finally, treatment of endothelial cells with HOCl stimulated mitochondrial dysfunction, caspase-3 activation, and cell death that was potentiated by the HO inhibitor, tin protoporphyrin-IX, or by the knockdown of HO-1, and reversed by the exogenous administration of biliverdin, bilirubin, or CO. Hypochlorous Acid 45-49 heme oxygenase 1 Homo sapiens 213-217 19625608-7 2009 Finally, treatment of endothelial cells with HOCl stimulated mitochondrial dysfunction, caspase-3 activation, and cell death that was potentiated by the HO inhibitor, tin protoporphyrin-IX, or by the knockdown of HO-1, and reversed by the exogenous administration of biliverdin, bilirubin, or CO. Biliverdine 267-277 heme oxygenase 1 Homo sapiens 213-217 19625608-7 2009 Finally, treatment of endothelial cells with HOCl stimulated mitochondrial dysfunction, caspase-3 activation, and cell death that was potentiated by the HO inhibitor, tin protoporphyrin-IX, or by the knockdown of HO-1, and reversed by the exogenous administration of biliverdin, bilirubin, or CO. Bilirubin 279-288 heme oxygenase 1 Homo sapiens 213-217 19625608-8 2009 These results demonstrate that HOCl induces HO-1 gene transcription via the activation of the Nrf2/ARE pathway to counteract HOCl-mediated mitochondrial dysfunction and cell death. Hypochlorous Acid 31-35 heme oxygenase 1 Homo sapiens 44-48 19625608-8 2009 These results demonstrate that HOCl induces HO-1 gene transcription via the activation of the Nrf2/ARE pathway to counteract HOCl-mediated mitochondrial dysfunction and cell death. Hypochlorous Acid 125-129 heme oxygenase 1 Homo sapiens 44-48 19625608-9 2009 The ability of HOCl to activate HO-1 gene expression may represent a critical adaptive response to maintain endothelial cell viability at sites of vascular inflammation and atherosclerosis. Hypochlorous Acid 15-19 heme oxygenase 1 Homo sapiens 32-36 19576919-0 2009 Methamphetamine induces heme oxygenase-1 expression in cortical neurons and glia to prevent its toxicity. Methamphetamine 0-15 heme oxygenase 1 Homo sapiens 24-40 19576919-3 2009 We used primary cortical neuron/glia cocultures to explore the role of HO-1 in METH-induced toxicity. Methamphetamine 79-83 heme oxygenase 1 Homo sapiens 71-75 19576919-8 2009 Inhibition of the p38 mitogen-activated protein kinase pathway significantly blocked HO-1 induction by METH and aggravated METH neurotoxicity. Methamphetamine 103-107 heme oxygenase 1 Homo sapiens 85-89 19576919-10 2009 However, prior induction of HO-1 using cobalt protoporphyrine IX partially protected neurons from METH toxicity. cobalt protoporphyrine ix 39-64 heme oxygenase 1 Homo sapiens 28-32 19576919-10 2009 However, prior induction of HO-1 using cobalt protoporphyrine IX partially protected neurons from METH toxicity. Methamphetamine 98-102 heme oxygenase 1 Homo sapiens 28-32 19576919-11 2009 Taken together, our results suggest that induction of HO-1 by METH via the p38 signaling pathway may be protective, albeit insufficient to completely protect cortical neurons from METH toxicity. Methamphetamine 62-66 heme oxygenase 1 Homo sapiens 54-58 19683516-6 2009 Depletion of glutathione in the cells or loading the cells with ascorbate greatly increased heme oxygenase-1 induction in the presence of copper. Glutathione 13-24 heme oxygenase 1 Homo sapiens 92-108 19683516-6 2009 Depletion of glutathione in the cells or loading the cells with ascorbate greatly increased heme oxygenase-1 induction in the presence of copper. Ascorbic Acid 64-73 heme oxygenase 1 Homo sapiens 92-108 19683516-6 2009 Depletion of glutathione in the cells or loading the cells with ascorbate greatly increased heme oxygenase-1 induction in the presence of copper. Copper 138-144 heme oxygenase 1 Homo sapiens 92-108 19855245-14 2009 Moreover, treatment with TAK-779 (a) decreased alloantigen-specific T-lymphocyte proliferation and number of IFN-gamma producing cells and (b) increased HO-1 gene transcript level in the allografts. TAK 779 25-32 heme oxygenase 1 Homo sapiens 153-157 19855245-17 2009 The beneficial effects of TAK-779 may be because of (a) reduction in CCR5 and CXCR3 T-lymphocyte subset infiltration into the graft, (b) attenuation of alloantigen-specific T-lymphocyte proliferative response and IFN-gamma production, and (c) induction of HO-1 gene. TAK 779 26-33 heme oxygenase 1 Homo sapiens 256-260 19808972-6 2009 The effect of ho-1 silencing on these oncogenic features was mimicked by exposure of cells to a novel selective small-molecule HO-1 inhibitor referred to as OB-24. OB-24 HCl 157-162 heme oxygenase 1 Homo sapiens 14-18 19808972-6 2009 The effect of ho-1 silencing on these oncogenic features was mimicked by exposure of cells to a novel selective small-molecule HO-1 inhibitor referred to as OB-24. OB-24 HCl 157-162 heme oxygenase 1 Homo sapiens 127-131 19808972-7 2009 OB-24 selectively inhibited HO-1 activity in PCA cells, which correlated with a reduction of protein carbonylation and reactive oxygen species formation. OB-24 HCl 0-5 heme oxygenase 1 Homo sapiens 28-32 19625608-3 2009 Treatment of human endothelial cells with the myeloperoxidase product HOCl stimulated a concentration- and time-dependent increase in HO-1 protein that resulted in a significant rise in carbon monoxide (CO) production. Hypochlorous Acid 70-74 heme oxygenase 1 Homo sapiens 134-138 19602432-2 2009 Silica nanoparticles-induced oxidative stress was assessed by examining the formation of reactive oxygen species (ROS) and induction of antioxidant enzymes, such as superoxide dismutase (SOD) and heme oxygenase-1 (HO-1). Silicon Dioxide 0-6 heme oxygenase 1 Homo sapiens 196-212 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Carbon Monoxide 102-117 heme oxygenase 1 Homo sapiens 18-22 19874266-5 2009 Induction of the astroglial ho-1 gene may constitute a "common pathway" leading to pathological brain iron deposition, intracellular oxidative damage and bioenergetic failure in AD and other human CNS disorders. Iron 102-106 heme oxygenase 1 Homo sapiens 28-32 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Heme 68-72 heme oxygenase 1 Homo sapiens 0-16 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Heme 68-72 heme oxygenase 1 Homo sapiens 18-22 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Biliverdine 76-86 heme oxygenase 1 Homo sapiens 0-16 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Biliverdine 76-86 heme oxygenase 1 Homo sapiens 18-22 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Iron 93-97 heme oxygenase 1 Homo sapiens 0-16 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Iron 93-97 heme oxygenase 1 Homo sapiens 18-22 19874266-2 2009 Heme oxygenase-1 (HO-1) is a 32 kDa stress protein that catabolizes heme to biliverdin, free iron and carbon monoxide. Carbon Monoxide 102-117 heme oxygenase 1 Homo sapiens 0-16 19364390-8 2009 Sublethal concentrations of arecoline upregulated the expression of the following stress-responsive genes: heme oxygenase-1; ferritin light chain; glucose-6-phosphate dehydrogenase; glutamate-cysteine ligase catalytic subunit; and glutathione reductase. Arecoline 28-37 heme oxygenase 1 Homo sapiens 107-123 19602432-2 2009 Silica nanoparticles-induced oxidative stress was assessed by examining the formation of reactive oxygen species (ROS) and induction of antioxidant enzymes, such as superoxide dismutase (SOD) and heme oxygenase-1 (HO-1). Silicon Dioxide 0-6 heme oxygenase 1 Homo sapiens 214-218 19602432-4 2009 From the overall results, silica nanoparticles exerted toxicity via oxidative stress, which lead to the induction of HO-1 via the Nrf-2-ERK MAP kinase signaling pathway; cells exposed to porous silica nanoparticles showed a more sensitive response than those exposed to fumed silica. Silicon Dioxide 26-32 heme oxygenase 1 Homo sapiens 117-121 19602432-4 2009 From the overall results, silica nanoparticles exerted toxicity via oxidative stress, which lead to the induction of HO-1 via the Nrf-2-ERK MAP kinase signaling pathway; cells exposed to porous silica nanoparticles showed a more sensitive response than those exposed to fumed silica. Silicon Dioxide 194-200 heme oxygenase 1 Homo sapiens 117-121 19602432-4 2009 From the overall results, silica nanoparticles exerted toxicity via oxidative stress, which lead to the induction of HO-1 via the Nrf-2-ERK MAP kinase signaling pathway; cells exposed to porous silica nanoparticles showed a more sensitive response than those exposed to fumed silica. Silicon Dioxide 194-200 heme oxygenase 1 Homo sapiens 117-121 19777608-0 2009 Iron increases HMOX1 and decreases hepatitis C viral expression in HCV-expressing cells. Iron 0-4 heme oxygenase 1 Homo sapiens 15-20 19540194-5 2009 C18-DOM increased the expression of anti-atherogenic molecule namely heme oxygenase-1 in endothelial cells and all these data showed that C18-DOM is exhibiting aspirin-like effects. Aspirin 160-167 heme oxygenase 1 Homo sapiens 69-85 19617398-1 2009 Heme oxygenase-1 (HO-1), a ubiquitous inducible stress-response protein, serves a major metabolic function in heme turnover. Heme 110-114 heme oxygenase 1 Homo sapiens 0-16 19617398-1 2009 Heme oxygenase-1 (HO-1), a ubiquitous inducible stress-response protein, serves a major metabolic function in heme turnover. Heme 110-114 heme oxygenase 1 Homo sapiens 18-22 19694439-2 2009 In humans, heme oxygenase-1 (hHO-1) is overexpressed in tumor tissues, where it helps to protect cancer cells from anticancer agents, while HOs in fungal pathogens, such as Candida albicans, function as the primary means of iron acquisition. Iron 224-228 heme oxygenase 1 Homo sapiens 11-27 19694439-2 2009 In humans, heme oxygenase-1 (hHO-1) is overexpressed in tumor tissues, where it helps to protect cancer cells from anticancer agents, while HOs in fungal pathogens, such as Candida albicans, function as the primary means of iron acquisition. Iron 224-228 heme oxygenase 1 Homo sapiens 29-34 19694439-4 2009 In this study, we have examined the equilibrium binding of three isocyanides, isopropyl, n-butyl, and benzyl, to the two major human HO isoforms (hHO-1 and hHO-2), Candida albicans HO (CaHmx1), and human cytochrome P450 CYP3A4 using electronic absorption spectroscopy. Cyanides 65-76 heme oxygenase 1 Homo sapiens 146-151 19694439-7 2009 Because the dissociation constants (KD) of the ligands for ferrous heme-hHO-1 were below the limit of accuracy for equilibrium titrations, stopped-flow kinetic experiments were used to measure the binding parameters of the isocyanides to ferrous hHO-1. Cyanides 223-234 heme oxygenase 1 Homo sapiens 72-77 19694439-7 2009 Because the dissociation constants (KD) of the ligands for ferrous heme-hHO-1 were below the limit of accuracy for equilibrium titrations, stopped-flow kinetic experiments were used to measure the binding parameters of the isocyanides to ferrous hHO-1. Cyanides 223-234 heme oxygenase 1 Homo sapiens 246-251 19694439-8 2009 Steady-state activity assays showed that benzyl isocyanide was the most potent uncompetitive inhibitor with respect to heme with a KI = 0.15 microM for hHO-1. Benzyl isocyanide 41-58 heme oxygenase 1 Homo sapiens 152-157 19694439-8 2009 Steady-state activity assays showed that benzyl isocyanide was the most potent uncompetitive inhibitor with respect to heme with a KI = 0.15 microM for hHO-1. Heme 119-123 heme oxygenase 1 Homo sapiens 152-157 19761582-13 2009 Ag-np induced stress resulted in the up regulation of metallothionein and heme oxygenase -1 genes. ag-np 0-5 heme oxygenase 1 Homo sapiens 74-91 19563780-2 2009 We identified the protein kinase C delta (PKCdelta) inhibitor rottlerin as the only compound that reduced NO-induced ARE-luciferase reporter activity and diminished NO-induced up-regulation of two Nrf2/ARE-regulated proteins - NAD(P)H:quinone oxidoreductase-1 (NQO1) and hemeoxygenase-1 (HO-1) in SH-Sy5y cells. rottlerin 62-71 heme oxygenase 1 Homo sapiens 271-292 19570893-0 2009 Membrane depolarization induces calcium-dependent upregulation of Hsp70 and Hmox-1 in skeletal muscle cells. Calcium 32-39 heme oxygenase 1 Homo sapiens 76-82 19570893-4 2009 The aim of this work was to investigate the involvement of calcium in Hsp70 and Hmox-1 expression upon depolarization of skeletal muscle cells. Calcium 59-66 heme oxygenase 1 Homo sapiens 80-86 19570893-8 2009 Here we demonstrated that inhibition of IP(3)-dependent calcium signals decreased both Hsp70 mRNA induction and Hsp70 and Hmox-1 protein expression. Inositol 1,4,5-Trisphosphate 40-45 heme oxygenase 1 Homo sapiens 122-128 19570893-8 2009 Here we demonstrated that inhibition of IP(3)-dependent calcium signals decreased both Hsp70 mRNA induction and Hsp70 and Hmox-1 protein expression. Calcium 56-63 heme oxygenase 1 Homo sapiens 122-128 19681826-7 2009 HO-1 inhibition by the specific inhibitor, SnPP, completely abolished T-cell suppression and IL-10 production. S-Nitroso-N-propionyl-D,L-penicillamine 43-47 heme oxygenase 1 Homo sapiens 0-4 19777608-4 2009 RESULTS: Iron, in the form of ferric nitrilotriacetate, increased oxidative stress and up-regulated HMOX1 gene expression. Iron 9-13 heme oxygenase 1 Homo sapiens 100-105 19777608-4 2009 RESULTS: Iron, in the form of ferric nitrilotriacetate, increased oxidative stress and up-regulated HMOX1 gene expression. ferric nitrilotriacetate 30-54 heme oxygenase 1 Homo sapiens 100-105 19777608-6 2009 Silencing the up-regulation of HMOX1 nuclear factor-erythroid 2-related factor 2 (Nrf2) by Nrf2-siRNA decreased FeNTA-mediated up-regulation of HMOX1 mRNA levels. ferric nitrilotriacetate 112-117 heme oxygenase 1 Homo sapiens 31-36 19777608-6 2009 Silencing the up-regulation of HMOX1 nuclear factor-erythroid 2-related factor 2 (Nrf2) by Nrf2-siRNA decreased FeNTA-mediated up-regulation of HMOX1 mRNA levels. ferric nitrilotriacetate 112-117 heme oxygenase 1 Homo sapiens 144-149 19777608-9 2009 CONCLUSION: Excess iron up-regulates HMOX1 and down-regulates HCV gene expression in hepatoma cells. Iron 19-23 heme oxygenase 1 Homo sapiens 37-42 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 40-44 heme oxygenase 1 Homo sapiens 0-16 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 40-44 heme oxygenase 1 Homo sapiens 18-22 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 101-105 heme oxygenase 1 Homo sapiens 0-16 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 101-105 heme oxygenase 1 Homo sapiens 18-22 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 101-105 heme oxygenase 1 Homo sapiens 0-16 18774956-1 2009 Heme oxygenase-1 (HO-1) contribution to iron homeostasis has been postulated, because it facilitates iron recycling by liberating iron mostly from heme catabolism. Iron 101-105 heme oxygenase 1 Homo sapiens 18-22 18774956-4 2009 Here we postulated that HO-1 is critical in the regulation of ferroportin, the major cellular iron exporter, and hepcidin, the key regulator of iron homeostasis central in the pathogenesis of anemia of inflammation. Iron 94-98 heme oxygenase 1 Homo sapiens 24-28 18774956-4 2009 Here we postulated that HO-1 is critical in the regulation of ferroportin, the major cellular iron exporter, and hepcidin, the key regulator of iron homeostasis central in the pathogenesis of anemia of inflammation. Iron 144-148 heme oxygenase 1 Homo sapiens 24-28 18774956-5 2009 Our current experiments in human THP-1 monocytic cells indicate a HO-1-induced iron-mediated surface-ferroportin expression, consistent with the role of HO-1 in iron recycling. Iron 79-83 heme oxygenase 1 Homo sapiens 66-70 18774956-5 2009 Our current experiments in human THP-1 monocytic cells indicate a HO-1-induced iron-mediated surface-ferroportin expression, consistent with the role of HO-1 in iron recycling. Iron 79-83 heme oxygenase 1 Homo sapiens 153-157 18774956-5 2009 Our current experiments in human THP-1 monocytic cells indicate a HO-1-induced iron-mediated surface-ferroportin expression, consistent with the role of HO-1 in iron recycling. Iron 161-165 heme oxygenase 1 Homo sapiens 66-70 18774956-5 2009 Our current experiments in human THP-1 monocytic cells indicate a HO-1-induced iron-mediated surface-ferroportin expression, consistent with the role of HO-1 in iron recycling. Iron 161-165 heme oxygenase 1 Homo sapiens 153-157 18774956-12 2009 This study therefore shows a crucial role for HO-1 in maintaining body iron balance. Iron 71-75 heme oxygenase 1 Homo sapiens 46-50 19794918-7 2009 The amount of heme oxygenase-1 (HO-1) protein in the intestinal mucosa was significantly increased by lansoprazole, but not by omeprazole. Lansoprazole 102-114 heme oxygenase 1 Homo sapiens 14-30 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Heme 245-249 heme oxygenase 1 Homo sapiens 0-16 19794918-7 2009 The amount of heme oxygenase-1 (HO-1) protein in the intestinal mucosa was significantly increased by lansoprazole, but not by omeprazole. Lansoprazole 102-114 heme oxygenase 1 Homo sapiens 32-36 19794918-8 2009 These results suggest that lansoprazole, but not omeprazole, ameliorates indomethacin-induced small intestinal ulceration through upregulation of HO-1/carbon monoxide. Lansoprazole 27-39 heme oxygenase 1 Homo sapiens 146-150 19653226-0 2009 Protective effect of sulforaphane on indomethacin-induced cytotoxicity via heme oxygenase-1 expression in human intestinal Int 407 cells. sulforaphane 21-33 heme oxygenase 1 Homo sapiens 75-91 19653226-0 2009 Protective effect of sulforaphane on indomethacin-induced cytotoxicity via heme oxygenase-1 expression in human intestinal Int 407 cells. Indomethacin 37-49 heme oxygenase 1 Homo sapiens 75-91 19653226-2 2009 In the present study, we investigated the effect of sulforaphane on the expression of heme oxygenase-1 (HO-1) in human intestinal Int 407 cells. sulforaphane 52-64 heme oxygenase 1 Homo sapiens 86-102 19653226-2 2009 In the present study, we investigated the effect of sulforaphane on the expression of heme oxygenase-1 (HO-1) in human intestinal Int 407 cells. sulforaphane 52-64 heme oxygenase 1 Homo sapiens 104-108 19653226-3 2009 RT-PCR and Western blot data revealed that sulforaphane induced an increase in HO-1 expression at the mRNA and protein levels, respectively. sulforaphane 43-55 heme oxygenase 1 Homo sapiens 79-83 19653226-5 2009 Actinomycin D (an RNA synthesis inhibitor) and cycloheximide (a protein synthesis inhibitor) inhibited sulforaphane-responsive HO-1 mRNA expression, indicating that sulforaphane is a requirement for transcription and de novo protein synthesis. Dactinomycin 0-13 heme oxygenase 1 Homo sapiens 127-131 19653226-5 2009 Actinomycin D (an RNA synthesis inhibitor) and cycloheximide (a protein synthesis inhibitor) inhibited sulforaphane-responsive HO-1 mRNA expression, indicating that sulforaphane is a requirement for transcription and de novo protein synthesis. Cycloheximide 47-60 heme oxygenase 1 Homo sapiens 127-131 19653226-5 2009 Actinomycin D (an RNA synthesis inhibitor) and cycloheximide (a protein synthesis inhibitor) inhibited sulforaphane-responsive HO-1 mRNA expression, indicating that sulforaphane is a requirement for transcription and de novo protein synthesis. sulforaphane 103-115 heme oxygenase 1 Homo sapiens 127-131 19653226-5 2009 Actinomycin D (an RNA synthesis inhibitor) and cycloheximide (a protein synthesis inhibitor) inhibited sulforaphane-responsive HO-1 mRNA expression, indicating that sulforaphane is a requirement for transcription and de novo protein synthesis. sulforaphane 165-177 heme oxygenase 1 Homo sapiens 127-131 19653226-7 2009 We also found that U0126, an ERK kinase inhibitor, suppressed the sulforaphane-induced HO-1 expression and nuclear translocation of Nrf2. U 0126 19-24 heme oxygenase 1 Homo sapiens 87-91 19653226-7 2009 We also found that U0126, an ERK kinase inhibitor, suppressed the sulforaphane-induced HO-1 expression and nuclear translocation of Nrf2. sulforaphane 66-78 heme oxygenase 1 Homo sapiens 87-91 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. sulforaphane 39-51 heme oxygenase 1 Homo sapiens 123-127 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. sulforaphane 39-51 heme oxygenase 1 Homo sapiens 248-252 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. Indomethacin 55-68 heme oxygenase 1 Homo sapiens 123-127 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. Indomethacin 55-68 heme oxygenase 1 Homo sapiens 248-252 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. sulforaphane 167-179 heme oxygenase 1 Homo sapiens 123-127 19653226-8 2009 Moreover, the cytoprotective effect of sulforaphane on indomethancin-induced cytotoxicity was partially blocked by ERK and HO-1 inhibitors, further demonstrating that sulforaphane attenuated oxidative stress through a pathway that involved ERK and HO-1. sulforaphane 167-179 heme oxygenase 1 Homo sapiens 248-252 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Biliverdine 212-222 heme oxygenase 1 Homo sapiens 0-16 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Biliverdine 212-222 heme oxygenase 1 Homo sapiens 18-22 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Bilirubin 296-305 heme oxygenase 1 Homo sapiens 0-16 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Bilirubin 296-305 heme oxygenase 1 Homo sapiens 18-22 19556236-1 2009 Heme oxygenase-1 (HO-1), a stress-inducible enzyme anchored in the endoplasmic reticulum (ER) by a single transmembrane segment (TMS) located at the C terminus, interacts with NADPH cytochrome P450 reductase and biliverdin reductase to catalyze heme degradation to biliverdin and its metabolite, bilirubin. Heme 245-249 heme oxygenase 1 Homo sapiens 18-22 19608869-3 2009 In this study, we report that HO-1 was induced in cultured human tracheal smooth muscle cells after either treatment with a potent inducer of HO-1 activity, cobalt protoporphyrin IX, or infection with a recombinant adenovirus that carries the human HO-1 gene. cobaltiprotoporphyrin 157-181 heme oxygenase 1 Homo sapiens 30-34 19781175-1 2009 OBJECTIVE: To study the protective effect of islet xenograft and its possible mechanism of high expression of heme oxygenase-1 (HO-1) in donor pancreas islet induced by cobalt protoporphyrin (CoPP). cobaltiprotoporphyrin 169-190 heme oxygenase 1 Homo sapiens 110-126 19781175-1 2009 OBJECTIVE: To study the protective effect of islet xenograft and its possible mechanism of high expression of heme oxygenase-1 (HO-1) in donor pancreas islet induced by cobalt protoporphyrin (CoPP). cobaltiprotoporphyrin 169-190 heme oxygenase 1 Homo sapiens 128-132 19781175-1 2009 OBJECTIVE: To study the protective effect of islet xenograft and its possible mechanism of high expression of heme oxygenase-1 (HO-1) in donor pancreas islet induced by cobalt protoporphyrin (CoPP). cobaltiprotoporphyrin 192-196 heme oxygenase 1 Homo sapiens 110-126 19781175-1 2009 OBJECTIVE: To study the protective effect of islet xenograft and its possible mechanism of high expression of heme oxygenase-1 (HO-1) in donor pancreas islet induced by cobalt protoporphyrin (CoPP). cobaltiprotoporphyrin 192-196 heme oxygenase 1 Homo sapiens 128-132 19534727-0 2009 Human haem oxygenase-1 induction by nitro-linoleic acid is mediated by cAMP, AP-1 and E-box response element interactions. 10-nitro-9,12-octadecadienoic acid 36-55 heme oxygenase 1 Homo sapiens 6-22 19534727-0 2009 Human haem oxygenase-1 induction by nitro-linoleic acid is mediated by cAMP, AP-1 and E-box response element interactions. Cyclic AMP 71-75 heme oxygenase 1 Homo sapiens 6-22 19534727-2 2009 LNO2 (nitrolinoleic acid) induces expression of HO-1 (haem oxygenase-1), an enzyme that catabolizes haem into products exhibiting potent anti-inflammatory properties. 10-Nitrolinoleic acid 0-4 heme oxygenase 1 Homo sapiens 48-70 19534727-2 2009 LNO2 (nitrolinoleic acid) induces expression of HO-1 (haem oxygenase-1), an enzyme that catabolizes haem into products exhibiting potent anti-inflammatory properties. 10-nitro-9,12-octadecadienoic acid 6-24 heme oxygenase 1 Homo sapiens 48-70 19534727-6 2009 Site-directed mutagenesis of a CRE (cAMP-response element) or of a downstream NF-E2/AP-1 (activating protein-1) element, individually, within this 500 bp region modestly reduced activation of the HO-1 promoter by LNO2. Cyclic AMP 36-40 heme oxygenase 1 Homo sapiens 196-200 19608869-6 2009 HO-1 overexpression also attenuated TNF-alpha-induced oxidative stress, which was abrogated in the presence of both the HO-1 inhibitor, zinc protoporphyrin IX, as well as a carbon monoxide scavenger. zinc protoporphyrin 136-158 heme oxygenase 1 Homo sapiens 0-4 19608869-6 2009 HO-1 overexpression also attenuated TNF-alpha-induced oxidative stress, which was abrogated in the presence of both the HO-1 inhibitor, zinc protoporphyrin IX, as well as a carbon monoxide scavenger. zinc protoporphyrin 136-158 heme oxygenase 1 Homo sapiens 120-124 19608869-6 2009 HO-1 overexpression also attenuated TNF-alpha-induced oxidative stress, which was abrogated in the presence of both the HO-1 inhibitor, zinc protoporphyrin IX, as well as a carbon monoxide scavenger. Carbon Monoxide 173-188 heme oxygenase 1 Homo sapiens 0-4 19608869-8 2009 These results suggest that HO-1 functions as a suppressor of TNF-alpha signaling, not only by inhibiting the expression of adhesion molecules and generation of interleukin-6, but also by diminishing intracellular reactive oxygen species production and nuclear factor-kappaB activation in both cultured human tracheal smooth muscle cells and the airways of mice. Reactive Oxygen Species 213-236 heme oxygenase 1 Homo sapiens 27-31 19508170-7 2009 Interestingly, several of the drug partners, when applied as single agents, induced the expression of Hsp32 in neoplastic cells, suggesting that synergistic effects resulted from SMA-ZnPP-induced ablation of a Hsp32-mediated survival-pathway that is otherwise used by tumor cells to escape drug-induced apoptosis. zinc protoporphyrin 183-187 heme oxygenase 1 Homo sapiens 102-107 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Heme 57-61 heme oxygenase 1 Homo sapiens 75-79 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Biliverdine 168-178 heme oxygenase 1 Homo sapiens 57-73 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Biliverdine 168-178 heme oxygenase 1 Homo sapiens 75-79 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Iron 185-189 heme oxygenase 1 Homo sapiens 57-73 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Iron 185-189 heme oxygenase 1 Homo sapiens 75-79 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 57-73 19727608-3 2009 One of the key components of cellular stress response is heme oxygenase-1 (HO-1), the rate limiting enzyme in the process of degrading potentially toxic free heme into biliverdin, free iron and carbon monoxide. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 75-79 19652369-0 2009 Nitric oxide modulates osteoblastic differentiation with heme oxygenase-1 via the mitogen activated protein kinase and nuclear factor-kappaB pathways in human periodontal ligament cells. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 57-73 19640789-3 2009 Heme oxygenase-1 (HO-1) is a key enzyme that is indispensable for the temporal and spatial regulation of host response and, together with its essential metabolite carbon monoxide (CO), is crucial for maintaining homeostasis, inhibition of inflammation and the preservation of function and life. Carbon Monoxide 163-178 heme oxygenase 1 Homo sapiens 0-16 19640789-3 2009 Heme oxygenase-1 (HO-1) is a key enzyme that is indispensable for the temporal and spatial regulation of host response and, together with its essential metabolite carbon monoxide (CO), is crucial for maintaining homeostasis, inhibition of inflammation and the preservation of function and life. Carbon Monoxide 163-178 heme oxygenase 1 Homo sapiens 18-22 19640789-3 2009 Heme oxygenase-1 (HO-1) is a key enzyme that is indispensable for the temporal and spatial regulation of host response and, together with its essential metabolite carbon monoxide (CO), is crucial for maintaining homeostasis, inhibition of inflammation and the preservation of function and life. Carbon Monoxide 180-182 heme oxygenase 1 Homo sapiens 0-16 19640789-3 2009 Heme oxygenase-1 (HO-1) is a key enzyme that is indispensable for the temporal and spatial regulation of host response and, together with its essential metabolite carbon monoxide (CO), is crucial for maintaining homeostasis, inhibition of inflammation and the preservation of function and life. Carbon Monoxide 180-182 heme oxygenase 1 Homo sapiens 18-22 19508170-7 2009 Interestingly, several of the drug partners, when applied as single agents, induced the expression of Hsp32 in neoplastic cells, suggesting that synergistic effects resulted from SMA-ZnPP-induced ablation of a Hsp32-mediated survival-pathway that is otherwise used by tumor cells to escape drug-induced apoptosis. zinc protoporphyrin 183-187 heme oxygenase 1 Homo sapiens 210-215 19522732-0 2009 Brain sterol dysregulation in sporadic AD and MCI: relationship to heme oxygenase-1. Sterols 6-12 heme oxygenase 1 Homo sapiens 67-83 18830972-0 2009 GSH-dependent iNOS and HO-1 mediated apoptosis of human Jurkat cells induced by nickel(II). Glutathione 0-3 heme oxygenase 1 Homo sapiens 23-27 18830972-0 2009 GSH-dependent iNOS and HO-1 mediated apoptosis of human Jurkat cells induced by nickel(II). Nickel(2+) 80-90 heme oxygenase 1 Homo sapiens 23-27 18830972-7 2009 A significant increase in HO-1 mRNA levels was detected in nickel treated cells. Nickel 59-65 heme oxygenase 1 Homo sapiens 26-30 18830972-9 2009 Expression changes of HO-1 and iNOS were markedly blocked when Jurkat cells were preincubated with NAC, suggesting that ROS resulted in HO-1 and iNOS dysfunction in Jurkat cells. Acetylcysteine 99-102 heme oxygenase 1 Homo sapiens 22-26 18830972-9 2009 Expression changes of HO-1 and iNOS were markedly blocked when Jurkat cells were preincubated with NAC, suggesting that ROS resulted in HO-1 and iNOS dysfunction in Jurkat cells. Acetylcysteine 99-102 heme oxygenase 1 Homo sapiens 136-140 18830972-9 2009 Expression changes of HO-1 and iNOS were markedly blocked when Jurkat cells were preincubated with NAC, suggesting that ROS resulted in HO-1 and iNOS dysfunction in Jurkat cells. Reactive Oxygen Species 120-123 heme oxygenase 1 Homo sapiens 22-26 18830972-9 2009 Expression changes of HO-1 and iNOS were markedly blocked when Jurkat cells were preincubated with NAC, suggesting that ROS resulted in HO-1 and iNOS dysfunction in Jurkat cells. Reactive Oxygen Species 120-123 heme oxygenase 1 Homo sapiens 136-140 18830972-10 2009 We supposed that the immune toxicity of nickel(II) was mainly due to GSH depletion and finally led to apoptosis, probably via changing the expression levels of HO-1 and iNOS in human T lymphocytes. Nickel 40-46 heme oxygenase 1 Homo sapiens 160-164 19522732-3 2009 We recently determined that HO-1 over-expression suppresses total CH levels by augmenting liver X receptor-mediated CH efflux and enhances oxysterol formation in cultured astroglia. Oxysterols 139-148 heme oxygenase 1 Homo sapiens 28-32 19522732-6 2009 The relationships of sterol/oxysterol levels to HO-1 protein expression and clinical/demographic variables were determined by multivariable regression and non-parametric statistical analyses. Sterols 21-27 heme oxygenase 1 Homo sapiens 48-52 19522732-6 2009 The relationships of sterol/oxysterol levels to HO-1 protein expression and clinical/demographic variables were determined by multivariable regression and non-parametric statistical analyses. Oxysterols 28-37 heme oxygenase 1 Homo sapiens 48-52 19401559-0 2009 Identification of crassin acetate as a new immunosuppressant triggering heme oxygenase-1 expression in dendritic cells. crassin acetate 18-33 heme oxygenase 1 Homo sapiens 72-88 19656028-5 2009 Nicotine induced the synthesis of the transcription factor NF-E2-related factor-2 (Nrf2) as well as a number of cellular antioxidants and phase II enzymes, such as heme oxygenase-1. Nicotine 0-8 heme oxygenase 1 Homo sapiens 164-180 20021945-6 2009 The relative survival rate of HepG2 cells under hypoxia was significantly decreased after the HO-1 protein overexpression was inhibited by ZnPPIX (P < 0.01). zinc protoporphyrin 139-145 heme oxygenase 1 Homo sapiens 94-98 20021945-7 2009 The total SOD activity of cells was significantly increased after cells were treated by hypoxia for 16 hours (P < 0.05), while decreased significantly by HO-1 inhibitor ZnPPIX treatment (P < 0.01). zinc protoporphyrin 172-178 heme oxygenase 1 Homo sapiens 157-161 20021945-9 2009 In addition, the HO-1 overexpression under hypoxia was decreased by HIF-1alpha inhibitor, while the HIF-1alpha expression level under hypoxia was not significantly changed after HO-1 expression was inhibited by ZnPPIX. zinc protoporphyrin 211-217 heme oxygenase 1 Homo sapiens 178-182 19457866-6 2009 Synergy was observed between laminar shear stress and atorvastatin, resulting in optimal expression of heme oxygenase-1 and resistance to oxidative stress, a response inhibited by heme oxygenase-1 small interfering RNA. Atorvastatin 54-66 heme oxygenase 1 Homo sapiens 103-119 19457866-6 2009 Synergy was observed between laminar shear stress and atorvastatin, resulting in optimal expression of heme oxygenase-1 and resistance to oxidative stress, a response inhibited by heme oxygenase-1 small interfering RNA. Atorvastatin 54-66 heme oxygenase 1 Homo sapiens 180-196 19457866-9 2009 In contrast, heme oxygenase-1 induction by atorvastatin in endothelial cells exposed to oscillatory flow was markedly attenuated. Atorvastatin 43-55 heme oxygenase 1 Homo sapiens 13-29 19457866-10 2009 We have identified a novel relationship between laminar shear stress and statins, demonstrating that atorvastatin-mediated heme oxygenase-1-dependent antioxidant effects are laminar shear stress-dependent, proving the principle that biomechanical signaling contributes significantly to endothelial responsiveness to pharmacological agents. Atorvastatin 101-113 heme oxygenase 1 Homo sapiens 123-139 19401559-4 2009 Treatment with CRA at nontoxic doses induced heme oxygenase-1 (HO-1) mRNA/protein expression and HO-1 enzymatic activity in DCs, suggesting a unique mechanism of action. crassin acetate 15-18 heme oxygenase 1 Homo sapiens 45-61 19401559-4 2009 Treatment with CRA at nontoxic doses induced heme oxygenase-1 (HO-1) mRNA/protein expression and HO-1 enzymatic activity in DCs, suggesting a unique mechanism of action. crassin acetate 15-18 heme oxygenase 1 Homo sapiens 63-67 19401559-4 2009 Treatment with CRA at nontoxic doses induced heme oxygenase-1 (HO-1) mRNA/protein expression and HO-1 enzymatic activity in DCs, suggesting a unique mechanism of action. crassin acetate 15-18 heme oxygenase 1 Homo sapiens 97-101 19457084-7 2009 Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. Deferoxamine 112-124 heme oxygenase 1 Homo sapiens 23-27 18619522-8 2009 Heme oxygenase 1 (HO1) is an important component of the system for mobilization of iron from macrophages. Iron 83-87 heme oxygenase 1 Homo sapiens 0-16 18619522-8 2009 Heme oxygenase 1 (HO1) is an important component of the system for mobilization of iron from macrophages. Iron 83-87 heme oxygenase 1 Homo sapiens 18-21 19702533-2 2009 Naturally occurring substances that upregulate the inducible isoform of HO (HO-1) have therefore been proposed as potential new drugs for the treatment of free radical-induced disease. Free Radicals 155-167 heme oxygenase 1 Homo sapiens 76-80 19453654-0 2009 High extracellular induced sputum haem oxygenase-1 in sarcoidosis and chronic beryllium disease. Beryllium 78-87 heme oxygenase 1 Homo sapiens 34-50 19362144-3 2009 HO-1 serves a vital metabolic function as the rate-limiting step in the heme degradation pathway and in the maintenance of iron homeostasis. Heme 72-76 heme oxygenase 1 Homo sapiens 0-4 19362144-3 2009 HO-1 serves a vital metabolic function as the rate-limiting step in the heme degradation pathway and in the maintenance of iron homeostasis. Iron 123-127 heme oxygenase 1 Homo sapiens 0-4 19362144-5 2009 The cytoprotective functions of HO-1 may be attributed to heme turnover, as well as to beneficial properties of its enzymatic reaction products: biliverdin-IXalpha, iron, and carbon monoxide (CO). Biliverdine 145-163 heme oxygenase 1 Homo sapiens 32-36 19362144-5 2009 The cytoprotective functions of HO-1 may be attributed to heme turnover, as well as to beneficial properties of its enzymatic reaction products: biliverdin-IXalpha, iron, and carbon monoxide (CO). Iron 165-169 heme oxygenase 1 Homo sapiens 32-36 19362144-5 2009 The cytoprotective functions of HO-1 may be attributed to heme turnover, as well as to beneficial properties of its enzymatic reaction products: biliverdin-IXalpha, iron, and carbon monoxide (CO). Carbon Monoxide 175-190 heme oxygenase 1 Homo sapiens 32-36 19362144-5 2009 The cytoprotective functions of HO-1 may be attributed to heme turnover, as well as to beneficial properties of its enzymatic reaction products: biliverdin-IXalpha, iron, and carbon monoxide (CO). Carbon Monoxide 192-194 heme oxygenase 1 Homo sapiens 32-36 19475336-3 2009 Therefore, it is tempting that the iron-releasing key enzyme in heme catabolism, heme oxygenase-1 (HO-1), may represent a candidate for a genetic susceptibility to PD. Iron 35-39 heme oxygenase 1 Homo sapiens 81-97 19475336-3 2009 Therefore, it is tempting that the iron-releasing key enzyme in heme catabolism, heme oxygenase-1 (HO-1), may represent a candidate for a genetic susceptibility to PD. Iron 35-39 heme oxygenase 1 Homo sapiens 99-103 19475336-3 2009 Therefore, it is tempting that the iron-releasing key enzyme in heme catabolism, heme oxygenase-1 (HO-1), may represent a candidate for a genetic susceptibility to PD. Heme 64-68 heme oxygenase 1 Homo sapiens 81-97 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 139-154 heme oxygenase 1 Homo sapiens 29-45 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 139-154 heme oxygenase 1 Homo sapiens 47-51 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 139-154 heme oxygenase 1 Homo sapiens 125-129 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 156-158 heme oxygenase 1 Homo sapiens 29-45 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 156-158 heme oxygenase 1 Homo sapiens 47-51 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Carbon Monoxide 156-158 heme oxygenase 1 Homo sapiens 125-129 19465554-1 2009 Given that the physiology of heme oxygenase-1 (HO-1) encompasses mitochondrial biogenesis, we tested the hypothesis that the HO-1 product, carbon monoxide (CO), activates mitochondrial biogenesis in skeletal muscle and enhances maximal oxygen uptake (Vo(2max)) in humans. Oxygen 34-40 heme oxygenase 1 Homo sapiens 47-51 19590705-0 2009 Taurine Chloramine Activates Nrf2, Increases HO-1 Expression and Protects Cells from Death Caused by Hydrogen Peroxide. N-chlorotaurine 0-18 heme oxygenase 1 Homo sapiens 45-49 19590705-5 2009 TauCl increased the expression of peroxiredoxin-1, thioredoxin-1 and heme oxygenase (HO)-1, the anti-oxidant enzymes normally induced by activation of NF-E2-related factor-2 (Nrf2). N-chlorotaurine 0-5 heme oxygenase 1 Homo sapiens 69-90 19590705-8 2009 Elevation of HO activity via induction of HO-1 expression within neighboring cells may provide protection from cytotoxicity caused by inflammatory oxidants like H(2)O(2). Water 161-166 heme oxygenase 1 Homo sapiens 42-46 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Heme 49-53 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Heme 49-53 heme oxygenase 1 Homo sapiens 18-22 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Iron 62-66 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Iron 62-66 heme oxygenase 1 Homo sapiens 18-22 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 18-22 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 18-22 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 18-22 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Bilirubin 105-114 heme oxygenase 1 Homo sapiens 0-16 19457084-3 2009 Heme oxygenase-1 (HO-1), an enzyme that converts heme to free iron, carbon monoxide (CO) and biliverdin (bilirubin precursor) is expressed in response to various stressors. Bilirubin 105-114 heme oxygenase 1 Homo sapiens 18-22 19457084-7 2009 Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. tin mesoporphyrin 75-92 heme oxygenase 1 Homo sapiens 23-27 19457084-7 2009 Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. Iron 98-102 heme oxygenase 1 Homo sapiens 23-27 19475336-3 2009 Therefore, it is tempting that the iron-releasing key enzyme in heme catabolism, heme oxygenase-1 (HO-1), may represent a candidate for a genetic susceptibility to PD. Heme 64-68 heme oxygenase 1 Homo sapiens 99-103 19457084-7 2009 Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. Cyclic GMP 155-159 heme oxygenase 1 Homo sapiens 23-27 19457084-7 2009 Likewise, treatment of HO-1 over-expressing cells with the HO-1 inhibitor, tin mesoporphyrin, the iron chelator deferoxamine or antagonist of CO-dependent cGMP activation, methylene blue, mitigated the HO-1-induced reduction in alpha-synuclein levels. Methylene Blue 172-186 heme oxygenase 1 Homo sapiens 23-27 19457084-8 2009 Furthermore, when HO-1 over-expressing cells were treated with the proteosome inhibitors, lactacystin and MG132, level of alpha-synuclein was almost completely restored. lactacystin 90-101 heme oxygenase 1 Homo sapiens 18-22 19457084-8 2009 Furthermore, when HO-1 over-expressing cells were treated with the proteosome inhibitors, lactacystin and MG132, level of alpha-synuclein was almost completely restored. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 106-111 heme oxygenase 1 Homo sapiens 18-22 19457088-7 2009 Heme-derived free ferrous iron, CO, and biliverdin/bilirubin are biologically active substances that have been shown to either ameliorate or exacerbate neural injury contingent upon specific disease models employed, the intensity and duration of HO-1 expression and the nature of the prevailing redox microenvironment. ferrous iron 18-30 heme oxygenase 1 Homo sapiens 246-250 19457088-7 2009 Heme-derived free ferrous iron, CO, and biliverdin/bilirubin are biologically active substances that have been shown to either ameliorate or exacerbate neural injury contingent upon specific disease models employed, the intensity and duration of HO-1 expression and the nature of the prevailing redox microenvironment. Biliverdine 40-50 heme oxygenase 1 Homo sapiens 246-250 19457088-7 2009 Heme-derived free ferrous iron, CO, and biliverdin/bilirubin are biologically active substances that have been shown to either ameliorate or exacerbate neural injury contingent upon specific disease models employed, the intensity and duration of HO-1 expression and the nature of the prevailing redox microenvironment. Bilirubin 51-60 heme oxygenase 1 Homo sapiens 246-250 19457088-8 2009 In "stressed" astroglia, HO-1 hyperactivity promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure amply documented in Alzheimer disease, Parkinson disease and other aging-related neurodegenerative disorders. Iron 100-104 heme oxygenase 1 Homo sapiens 25-29 19457088-8 2009 In "stressed" astroglia, HO-1 hyperactivity promotes mitochondrial sequestration of non-transferrin iron and macroautophagy and may thereby contribute to the pathological iron deposition and bioenergetic failure amply documented in Alzheimer disease, Parkinson disease and other aging-related neurodegenerative disorders. Iron 171-175 heme oxygenase 1 Homo sapiens 25-29 19457088-9 2009 Glial HO-1 expression may also impact cell survival and neuroplasticity in these conditions by modulating brain sterol metabolism and proteosomal degradation of neurotoxic protein aggregates. Sterols 112-118 heme oxygenase 1 Homo sapiens 6-10 19039664-7 2009 The regulation of both hmox1 and mt1 is prevented by the copper-chelator, bathocuproinedisulfonate (BCDS), but not uptake of heme-AlexaFluor-labeled HPX into endosomes. bathocuproine sulfonate 74-98 heme oxygenase 1 Homo sapiens 23-28 19320847-5 2009 Targeted knockdown of HO-1 leads to decreased intracellular PpIX accumulation, resulting in a failure to enhance ALA-PDT effect in four cell lines. protoporphyrin IX 60-64 heme oxygenase 1 Homo sapiens 22-26 19320847-8 2009 Our results suggested that (1) FECH siRNA improved the phototoxicity of ALA-PDT, (2) overexpression of HO-1 was associated with shorter (GT)n repeat of the promoter region, and (3) siRNA-mediated knockdown of HO-1 could suppress the growth of bladder cancer cells overexpressing HO-1. Alanine 72-75 heme oxygenase 1 Homo sapiens 103-107 19320847-8 2009 Our results suggested that (1) FECH siRNA improved the phototoxicity of ALA-PDT, (2) overexpression of HO-1 was associated with shorter (GT)n repeat of the promoter region, and (3) siRNA-mediated knockdown of HO-1 could suppress the growth of bladder cancer cells overexpressing HO-1. Alanine 72-75 heme oxygenase 1 Homo sapiens 209-213 19320847-8 2009 Our results suggested that (1) FECH siRNA improved the phototoxicity of ALA-PDT, (2) overexpression of HO-1 was associated with shorter (GT)n repeat of the promoter region, and (3) siRNA-mediated knockdown of HO-1 could suppress the growth of bladder cancer cells overexpressing HO-1. Alanine 72-75 heme oxygenase 1 Homo sapiens 209-213 19428335-0 2009 Heme oxygenase-1 induction modulates microsomal prostaglandin E synthase-1 expression and prostaglandin E(2) production in osteoarthritic chondrocytes. Dinoprostone 90-108 heme oxygenase 1 Homo sapiens 0-16 19428335-5 2009 Cells were stimulated with IL-1beta in the absence or presence of the HO-1 inducer cobalt protoporphyrin IX (CoPP). cobaltiprotoporphyrin 83-107 heme oxygenase 1 Homo sapiens 70-74 19428335-9 2009 The production of PGE(2) was decreased by HO-1 induction as a result of diminished mPGES-1 protein and mRNA expression. Dinoprostone 18-24 heme oxygenase 1 Homo sapiens 42-46 19508729-6 2009 HO-1 expression and activity was regulated by zinc (inhibition) and cobalt (induction) protoporphyrin. Cobalt 68-74 heme oxygenase 1 Homo sapiens 0-4 19508729-6 2009 HO-1 expression and activity was regulated by zinc (inhibition) and cobalt (induction) protoporphyrin. protoporphyrin IX 85-101 heme oxygenase 1 Homo sapiens 0-4 19508729-7 2009 Furthermore, the influence of cellular HO-1 levels and its metabolites on effects of standard chemotherapy with gemcitabine was tested in vivo and in vitro. gemcitabine 112-123 heme oxygenase 1 Homo sapiens 39-43 19508729-9 2009 Chemoresistance to gemcitabine was increased during HO-1 induction in PaCa cells expressing low levels of HO-1. gemcitabine 19-30 heme oxygenase 1 Homo sapiens 52-56 19508729-9 2009 Chemoresistance to gemcitabine was increased during HO-1 induction in PaCa cells expressing low levels of HO-1. gemcitabine 19-30 heme oxygenase 1 Homo sapiens 106-110 19508729-14 2009 The metabolites biliverdin and iron seem to be involved in HO-1-mediated resistance to anticancer treatment. Biliverdine 16-26 heme oxygenase 1 Homo sapiens 59-63 19508729-14 2009 The metabolites biliverdin and iron seem to be involved in HO-1-mediated resistance to anticancer treatment. Iron 31-35 heme oxygenase 1 Homo sapiens 59-63 19344704-4 2009 Whereas the expression of glutathione peroxidase (GPx), catalase, Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and heme oxygenase-1 (HO-1) increased with curcumin concentration and also with increase in time of incubation, the expression of Mn- superoxide dismutase (Mn-SOD) showed concentration dependant repression upon treatment with curcumin. Curcumin 148-156 heme oxygenase 1 Homo sapiens 109-125 19344704-4 2009 Whereas the expression of glutathione peroxidase (GPx), catalase, Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and heme oxygenase-1 (HO-1) increased with curcumin concentration and also with increase in time of incubation, the expression of Mn- superoxide dismutase (Mn-SOD) showed concentration dependant repression upon treatment with curcumin. Curcumin 148-156 heme oxygenase 1 Homo sapiens 127-131 19487598-8 2009 HO-1 induction by cobalt protoporphyrin impeded lesion progression into vulnerable plaques, indicated by a reduction in necrotic core size and intraplaque lipid accumulation, whereas cap thickness and vascular smooth muscle cells were increased. cobaltiprotoporphyrin 18-39 heme oxygenase 1 Homo sapiens 0-4 19487598-9 2009 In contrast, inhibition of HO-1 by zinc protoporphyrin augmented plaque vulnerability. zinc protoporphyrin 35-54 heme oxygenase 1 Homo sapiens 27-31 19325051-0 2009 The protective role of HO-1 and its generated products (CO, bilirubin, and Fe) in ethanol-induced human hepatocyte damage. Carbon Monoxide 56-58 heme oxygenase 1 Homo sapiens 23-27 19325051-0 2009 The protective role of HO-1 and its generated products (CO, bilirubin, and Fe) in ethanol-induced human hepatocyte damage. Ethanol 82-89 heme oxygenase 1 Homo sapiens 23-27 19325051-1 2009 It has been reported that naturally occurring quercetin exerts hepatoprotective effects through heme oxygenase-1 (HO-1) induction. Quercetin 46-55 heme oxygenase 1 Homo sapiens 96-112 19325051-1 2009 It has been reported that naturally occurring quercetin exerts hepatoprotective effects through heme oxygenase-1 (HO-1) induction. Quercetin 46-55 heme oxygenase 1 Homo sapiens 114-118 19325051-2 2009 However, the precise mechanism of how ethanol-associated liver damage is counteracted by quercetin-enhanced HO-1 metabolism still remains unclear. Quercetin 89-98 heme oxygenase 1 Homo sapiens 108-112 19325051-9 2009 These results suggested that quercetin virtually attenuated ethanol-derived oxidative damage via HO-1 induction. Quercetin 29-38 heme oxygenase 1 Homo sapiens 97-101 19325051-9 2009 These results suggested that quercetin virtually attenuated ethanol-derived oxidative damage via HO-1 induction. Ethanol 60-67 heme oxygenase 1 Homo sapiens 97-101 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 0-4 heme oxygenase 1 Homo sapiens 71-87 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 0-4 heme oxygenase 1 Homo sapiens 89-94 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 52-56 heme oxygenase 1 Homo sapiens 71-87 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 52-56 heme oxygenase 1 Homo sapiens 89-94 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Iron 114-118 heme oxygenase 1 Homo sapiens 71-87 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Iron 114-118 heme oxygenase 1 Homo sapiens 89-94 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 52-56 heme oxygenase 1 Homo sapiens 71-87 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Heme 52-56 heme oxygenase 1 Homo sapiens 89-94 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Iron 168-172 heme oxygenase 1 Homo sapiens 71-87 19039664-2 2009 Heme uptake via endocytosis of heme-HPX followed by heme catabolism by heme oxygenase-1 (HMOX1) raises regulatory iron pools, thus linking heme metabolism with that of iron. Iron 168-172 heme oxygenase 1 Homo sapiens 89-94 19039664-4 2009 When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Copper 33-39 heme oxygenase 1 Homo sapiens 22-27 19039664-4 2009 When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Copper 100-106 heme oxygenase 1 Homo sapiens 22-27 19039664-4 2009 When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Copper 100-106 heme oxygenase 1 Homo sapiens 22-27 19039664-4 2009 When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Copper 100-106 heme oxygenase 1 Homo sapiens 22-27 19039664-4 2009 When heme-HPX induces HMOX1, the copper-storing metallothioneins (MTs) are also induced whereas the copper-responsive copper chaperone that delivers copper to Cu, Zn superoxide dismutase, CCS1, is decreased; both are known responses when cellular copper levels rise. Copper 100-106 heme oxygenase 1 Homo sapiens 22-27 19039664-7 2009 The regulation of both hmox1 and mt1 is prevented by the copper-chelator, bathocuproinedisulfonate (BCDS), but not uptake of heme-AlexaFluor-labeled HPX into endosomes. bathocuproine sulfonate 100-104 heme oxygenase 1 Homo sapiens 23-28 19039664-5 2009 Endocytosis of heme-HPX is needed to regulate CCS1 since the signaling ligand cobalt-protoporphyrin (CoPP)-HPX, which does not induce HMOX1 but does co-localize with heme-HPX in endosomes, also decreased CCS1. heme-hpx 15-23 heme oxygenase 1 Homo sapiens 134-139 19039664-7 2009 The regulation of both hmox1 and mt1 is prevented by the copper-chelator, bathocuproinedisulfonate (BCDS), but not uptake of heme-AlexaFluor-labeled HPX into endosomes. Copper 57-63 heme oxygenase 1 Homo sapiens 23-28 19039664-8 2009 Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. Copper 22-28 heme oxygenase 1 Homo sapiens 32-37 19039664-8 2009 Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. Copper 22-28 heme oxygenase 1 Homo sapiens 157-162 19039664-8 2009 Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. tetraethylenepentamine 105-128 heme oxygenase 1 Homo sapiens 157-162 19039664-8 2009 Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. tetraamine 136-146 heme oxygenase 1 Homo sapiens 32-37 19039664-8 2009 Supporting a role for copper in HMOX1 regulation by heme-HPX, nutritional copper deficiency generated by tetraethylene pentamine or 232 tetraamine prevented HMOX1 induction. tetraamine 136-146 heme oxygenase 1 Homo sapiens 157-162 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Copper 30-36 heme oxygenase 1 Homo sapiens 178-183 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Copper 30-36 heme oxygenase 1 Homo sapiens 287-292 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Heme 71-75 heme oxygenase 1 Homo sapiens 178-183 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Heme 71-75 heme oxygenase 1 Homo sapiens 287-292 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Heme 111-115 heme oxygenase 1 Homo sapiens 178-183 19039664-10 2009 A model is presented in which copper endocytosis together with that of heme-HPX provides a means to facilitate heme export from HPX in the maturing endosomes: heme is needed for hmox1 transcription, while cytosolic copper and CCS1 provide a link for the known simultaneous regulation of hmox1 and mt1 by heme-HPX. Heme 111-115 heme oxygenase 1 Homo sapiens 287-292 19063990-0 2009 Heme oxygenase-1 induction prevents neuronal damage triggered during mitochondrial inhibition: role of CO and bilirubin. Carbon Monoxide 103-105 heme oxygenase 1 Homo sapiens 0-16 19373083-7 2009 SUMMARY: The development of derivatives of probucol targeting anti-inflammatory and antioxidant processes, perhaps via induction of heme oxygenase-1, may add to the armamentarium of current agents used in treatment of atherosclerotic disease and diabetes. Probucol 43-51 heme oxygenase 1 Homo sapiens 132-148 19063990-0 2009 Heme oxygenase-1 induction prevents neuronal damage triggered during mitochondrial inhibition: role of CO and bilirubin. Bilirubin 110-119 heme oxygenase 1 Homo sapiens 0-16 19063990-2 2009 A protective role of the inducible isoform, HO-1, has been described in pathological conditions associated with reactive oxygen species (ROS) and oxidative damage. Reactive Oxygen Species 112-135 heme oxygenase 1 Homo sapiens 44-48 19063990-2 2009 A protective role of the inducible isoform, HO-1, has been described in pathological conditions associated with reactive oxygen species (ROS) and oxidative damage. Reactive Oxygen Species 137-140 heme oxygenase 1 Homo sapiens 44-48 19063990-3 2009 The aim of this study was to investigate the role of HO-1 in the neurotoxicity induced by the mitochondrial toxin 3-nitropropionic acid (3-NP) in primary cultures of cerebellar granule neurons (CGNs). 3-nitropropionic acid 114-135 heme oxygenase 1 Homo sapiens 53-57 19063990-3 2009 The aim of this study was to investigate the role of HO-1 in the neurotoxicity induced by the mitochondrial toxin 3-nitropropionic acid (3-NP) in primary cultures of cerebellar granule neurons (CGNs). 3-nitropropionic acid 137-141 heme oxygenase 1 Homo sapiens 53-57 19063990-6 2009 In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. cobaltiprotoporphyrin 59-80 heme oxygenase 1 Homo sapiens 13-17 19063990-6 2009 In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. cobaltiprotoporphyrin 82-86 heme oxygenase 1 Homo sapiens 13-17 19063990-6 2009 In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. 3-nitropropionic acid 115-119 heme oxygenase 1 Homo sapiens 13-17 19063990-6 2009 In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. Reactive Oxygen Species 166-169 heme oxygenase 1 Homo sapiens 13-17 19063990-6 2009 In addition, HO-1 up-regulation induced by the exposure to cobalt protoporphyrin (CoPP) before the incubation with 3-NP, prevented the cell death and the increase in ROS induced by 3-NP. 3-nitropropionic acid 181-185 heme oxygenase 1 Homo sapiens 13-17 19420259-5 2009 In Tsc2-deficient neurons, the expression of stress markers such as CHOP and HO-1 is increased, and this increase is completely reversed by the mTOR inhibitor rapamycin both in vitro and in vivo. Sirolimus 159-168 heme oxygenase 1 Homo sapiens 77-81 19350554-7 2009 In contrast, N-acetylcysteine, a potent cysteine reductive compound, significantly prevents up-regulation of HMOX1, GCLM, and CXCL2 genes, and repression of MMP9 and CCL22 genes induced by As(2)O(3). Acetylcysteine 13-29 heme oxygenase 1 Homo sapiens 109-114 19350554-7 2009 In contrast, N-acetylcysteine, a potent cysteine reductive compound, significantly prevents up-regulation of HMOX1, GCLM, and CXCL2 genes, and repression of MMP9 and CCL22 genes induced by As(2)O(3). Cysteine 21-29 heme oxygenase 1 Homo sapiens 109-114 19121922-1 2009 OBJECTIVE: We tested the hypothesis that lycopene supplementation reduces the expression of oxidant-responsive heme oxygenase-1 (HO-1) in basal conditions and in response to an oxidant challenge and determined whether this is temporally associated with increased cell viability. Lycopene 41-49 heme oxygenase 1 Homo sapiens 111-127 19121922-1 2009 OBJECTIVE: We tested the hypothesis that lycopene supplementation reduces the expression of oxidant-responsive heme oxygenase-1 (HO-1) in basal conditions and in response to an oxidant challenge and determined whether this is temporally associated with increased cell viability. Lycopene 41-49 heme oxygenase 1 Homo sapiens 129-133 19121922-2 2009 METHODS: We determined basal and stimulated ex vivo expression of HO-1 and cell viability in lymphocytes from volunteers after lycopene supplementation. Lycopene 127-135 heme oxygenase 1 Homo sapiens 66-70 19484149-10 2009 In contrast to annexin A11, HMOX1 immunoreactivity positively correlated with in vitro cisplatin resistance in ovarian cancers. Cisplatin 87-96 heme oxygenase 1 Homo sapiens 28-33 19429248-3 2009 The overall results suggest that CeO(2) nanoparticles may exert their toxicity through oxidative stress, as they cause significant increases in the cellular reactive oxygen species (ROS) concentrations, subsequently leading to the strong induction of heme oxygenase-1 (HO-1) via the p38-Nrf-2 signaling pathway. Reactive Oxygen Species 157-180 heme oxygenase 1 Homo sapiens 251-267 19429248-3 2009 The overall results suggest that CeO(2) nanoparticles may exert their toxicity through oxidative stress, as they cause significant increases in the cellular reactive oxygen species (ROS) concentrations, subsequently leading to the strong induction of heme oxygenase-1 (HO-1) via the p38-Nrf-2 signaling pathway. Reactive Oxygen Species 182-185 heme oxygenase 1 Homo sapiens 251-267 19349368-5 2009 Incubating human microvascular endothelial cells with the sphingosine-1-phosphate type 1 receptor (S1P(1)) inhibitor VPC23019 or performing small interfering RNA knockdown of S1P(1) blocked arsenic-stimulated HMVEC angiogenic gene expression and tube formation, but did not affect induction of either HMOX1 or IL8. Arsenic 190-197 heme oxygenase 1 Homo sapiens 301-306 19251588-9 2009 In contrast, heme oxygenase-1 was strongly suppressed by DFO, both in the absence and presence of LPS or iron. Deferoxamine 57-60 heme oxygenase 1 Homo sapiens 13-29 19251588-9 2009 In contrast, heme oxygenase-1 was strongly suppressed by DFO, both in the absence and presence of LPS or iron. Iron 105-109 heme oxygenase 1 Homo sapiens 13-29 19384082-2 2009 Excess free heme catalyzes the formation of reactive oxygen species, which leads to endothelial cell (EC) dysfunction as seen in numerous pathologic vascular conditions including systemic hypertension and diabetes, as well as in ischemia/reperfusion injury.The up-regulation of HO-1 can be achieved through the use of pharmaceutical agents such as metalloporphyrins and statins. Reactive Oxygen Species 44-67 heme oxygenase 1 Homo sapiens 278-282 19384082-3 2009 In addition, atrial natriuretic peptide and nitric oxide donors are important modulators of the heme-HO system, either through induction of HO-1 or the increased biologic activity of its products. Nitric Oxide 44-56 heme oxygenase 1 Homo sapiens 140-144 19384082-6 2009 Delivery of human HO-1 to hyperglycemic rats significantly lowers superoxide levels and prevents EC damage and sloughing of vascular EC into the circulation. Superoxides 66-76 heme oxygenase 1 Homo sapiens 18-22 19244544-2 2009 Since HO-1 exerts potent antioxidant effects, we hypothesized that this enzyme inhibits ROS-induced cardiomyocyte hypertrophy. Reactive Oxygen Species 88-91 heme oxygenase 1 Homo sapiens 6-10 19117929-3 2009 METHODS: The ability of eriodictyol to activate Nrf2 and to induce the phase 2 proteins heme-oxygenase (HO)-1 and NAD(P)H:quinone oxidoreductase (NQO)-1, and the cellular antioxidant glutathione (GSH) were analyzed. eriodictyol 24-35 heme oxygenase 1 Homo sapiens 88-109 19245827-10 2009 In summary, these findings suggest that induction of HO-1 through a Smad-dependent mechanism is responsible for the cytoprotective effect of BMP6 in H(2)O(2)-mediated renal cell injury. Hydrogen Peroxide 149-157 heme oxygenase 1 Homo sapiens 53-57 19526463-0 2009 Investigations of the posttranslational mechanism of arsenite-mediated downregulation of human cytochrome P4501A1 levels: the role of heme oxygenase-1. arsenite 53-61 heme oxygenase 1 Homo sapiens 134-150 19526463-3 2009 We hypothesize that arsenite induces heme oxygenase-1 (HO-1), which catabolizes CYP1A1 heme or cellular heme pools, thereby downregulating CYP1A1. arsenite 20-28 heme oxygenase 1 Homo sapiens 37-53 19526463-7 2009 Together these findings demonstrate that a posttranslational mechanism involving decreases in the cellular heme pool by arsenite-induced HO-1 may contribute to arsenite-mediated downregulation of CYP1A1. arsenite 160-168 heme oxygenase 1 Homo sapiens 137-141 19526463-3 2009 We hypothesize that arsenite induces heme oxygenase-1 (HO-1), which catabolizes CYP1A1 heme or cellular heme pools, thereby downregulating CYP1A1. arsenite 20-28 heme oxygenase 1 Homo sapiens 55-59 19526463-3 2009 We hypothesize that arsenite induces heme oxygenase-1 (HO-1), which catabolizes CYP1A1 heme or cellular heme pools, thereby downregulating CYP1A1. Heme 37-41 heme oxygenase 1 Homo sapiens 55-59 19526463-3 2009 We hypothesize that arsenite induces heme oxygenase-1 (HO-1), which catabolizes CYP1A1 heme or cellular heme pools, thereby downregulating CYP1A1. Heme 87-91 heme oxygenase 1 Homo sapiens 37-53 19526463-3 2009 We hypothesize that arsenite induces heme oxygenase-1 (HO-1), which catabolizes CYP1A1 heme or cellular heme pools, thereby downregulating CYP1A1. Heme 87-91 heme oxygenase 1 Homo sapiens 55-59 19526463-4 2009 Arsenite (5 microM), in HepG2 cells, induced HO-1 mRNA 7.4-fold over the 48 h observation period, and it upregulated HO-1 protein expression. arsenite 0-8 heme oxygenase 1 Homo sapiens 45-49 19526463-4 2009 Arsenite (5 microM), in HepG2 cells, induced HO-1 mRNA 7.4-fold over the 48 h observation period, and it upregulated HO-1 protein expression. arsenite 0-8 heme oxygenase 1 Homo sapiens 117-121 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. arsenite 0-8 heme oxygenase 1 Homo sapiens 152-156 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. Polycyclic Aromatic Hydrocarbons 48-51 heme oxygenase 1 Homo sapiens 152-156 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. benzo(k)fluoranthene 53-73 heme oxygenase 1 Homo sapiens 152-156 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. benzo(k)fluoranthene 75-78 heme oxygenase 1 Homo sapiens 152-156 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. arsenite 175-183 heme oxygenase 1 Homo sapiens 152-156 19526463-5 2009 Arsenite decreased the induction of CYP1A1 by a PAH, benzo[k]fluoranthene (BKF), by 50%; and transfection of HepG2 cells with siRNA targeting the human HO-1 gene, reduced the arsenite downregulation of BKF-induced CYP1A1 from 54% to 27%, relative to untransfected cells. benzo(k)fluoranthene 202-205 heme oxygenase 1 Homo sapiens 152-156 19526463-7 2009 Together these findings demonstrate that a posttranslational mechanism involving decreases in the cellular heme pool by arsenite-induced HO-1 may contribute to arsenite-mediated downregulation of CYP1A1. Heme 107-111 heme oxygenase 1 Homo sapiens 137-141 19526463-7 2009 Together these findings demonstrate that a posttranslational mechanism involving decreases in the cellular heme pool by arsenite-induced HO-1 may contribute to arsenite-mediated downregulation of CYP1A1. arsenite 120-128 heme oxygenase 1 Homo sapiens 137-141 19250338-0 2009 HO-1-mediated macroautophagy: a mechanism for unregulated iron deposition in aging and degenerating neural tissues. Iron 58-62 heme oxygenase 1 Homo sapiens 0-4 19250338-2 2009 We previously demonstrated that heme oxygenase-1 (HO-1) is up-regulated in AD and PD brain and promotes the accumulation of non-transferrin iron in astroglial mitochondria. Iron 140-144 heme oxygenase 1 Homo sapiens 32-48 19250338-6 2009 Thus, HO-1 activity promotes mitochondrial macroautophagy and sequestration of redox-active iron in astroglia independently of classical iron mobilization pathways. Iron 92-96 heme oxygenase 1 Homo sapiens 6-10 19250338-6 2009 Thus, HO-1 activity promotes mitochondrial macroautophagy and sequestration of redox-active iron in astroglia independently of classical iron mobilization pathways. Iron 137-141 heme oxygenase 1 Homo sapiens 6-10 19250338-7 2009 Glial HO-1 may be a rational therapeutic target in AD, PD, and other human CNS conditions characterized by the unregulated deposition of brain iron. Iron 143-147 heme oxygenase 1 Homo sapiens 6-10 19250338-2 2009 We previously demonstrated that heme oxygenase-1 (HO-1) is up-regulated in AD and PD brain and promotes the accumulation of non-transferrin iron in astroglial mitochondria. Iron 140-144 heme oxygenase 1 Homo sapiens 50-54 19250338-5 2009 HO-1 promoted trapping of redox-active iron and sulfur within many cytopathological profiles without impacting ferroportin, transferrin receptor, ferritin, and IRP2 protein levels or IRP1 activity. Iron 39-43 heme oxygenase 1 Homo sapiens 0-4 19250338-5 2009 HO-1 promoted trapping of redox-active iron and sulfur within many cytopathological profiles without impacting ferroportin, transferrin receptor, ferritin, and IRP2 protein levels or IRP1 activity. Sulfur 48-54 heme oxygenase 1 Homo sapiens 0-4 19289500-0 2009 Analysis of nitric oxide-stabilized mRNAs in human fibroblasts reveals HuR-dependent heme oxygenase 1 upregulation. Nitric Oxide 12-24 heme oxygenase 1 Homo sapiens 85-101 19289500-1 2009 We previously observed that nitric oxide (NO) exposure increases the stability of mRNAs encoding heme oxygenase 1 (HO-1) and TIEG-1 in human and mouse fibroblasts. Nitric Oxide 28-40 heme oxygenase 1 Homo sapiens 97-113 19289500-1 2009 We previously observed that nitric oxide (NO) exposure increases the stability of mRNAs encoding heme oxygenase 1 (HO-1) and TIEG-1 in human and mouse fibroblasts. Nitric Oxide 28-40 heme oxygenase 1 Homo sapiens 115-119 19323979-5 2009 Treatment of HUVEC with ATN-224 caused increased superoxide levels, phospho-ERK signalling, nuclear NRF1 expression, HO-1 expression and induction of the anti-apoptotic proteins P21, BCL2 and BCLXL. tetrathiomolybdate 24-31 heme oxygenase 1 Homo sapiens 117-121 19388153-9 2009 Arecoline was also found to elevate HO-1 mRNA and protein expression in a dose-dependent manner (P < 0.05). Arecoline 0-9 heme oxygenase 1 Homo sapiens 36-40 19388153-10 2009 CONCLUSIONS: Taken together, the data presented here demonstrated that HO-1 expression is significantly upregulated in OSF from areca quid chewers, and arecoline may be responsible for the enhanced HO-1 expression in vivo. Arecoline 152-161 heme oxygenase 1 Homo sapiens 198-202 18485456-0 2009 Upregulation of heme oxygenase-1 as a host mechanism for protection against nitric oxide-induced damage in human renal epithelial cells. Nitric Oxide 76-88 heme oxygenase 1 Homo sapiens 16-32 19651797-5 2009 Two electrophiles, 4-hydroxynonenal (HNE) and acrolein, induced the expression of phase II genes (GCLC, GCLM, NQO1, NQO2, HO-1, and GSTM-1). 4-hydroxy-2-nonenal 19-35 heme oxygenase 1 Homo sapiens 122-126 19651797-5 2009 Two electrophiles, 4-hydroxynonenal (HNE) and acrolein, induced the expression of phase II genes (GCLC, GCLM, NQO1, NQO2, HO-1, and GSTM-1). Acrolein 46-54 heme oxygenase 1 Homo sapiens 122-126 18485456-2 2009 METHODS: Heme oxygenase-1 expression was investigated in the human renal epithelial cell line A498 in response to the uropathogenic Escherichia coli (UPEC) strain IA2, the NO-donor DETA/NONOate (DETA/NO), and proinflammatory cytokines (interleukin-1beta, tumor necrosis factor-alpha, and interferon-gamma) using reverse transcriptase polymerase chain reaction and Western blot analysis. DEET 181-185 heme oxygenase 1 Homo sapiens 9-25 18485456-2 2009 METHODS: Heme oxygenase-1 expression was investigated in the human renal epithelial cell line A498 in response to the uropathogenic Escherichia coli (UPEC) strain IA2, the NO-donor DETA/NONOate (DETA/NO), and proinflammatory cytokines (interleukin-1beta, tumor necrosis factor-alpha, and interferon-gamma) using reverse transcriptase polymerase chain reaction and Western blot analysis. pelargonic acid 186-193 heme oxygenase 1 Homo sapiens 9-25 19243105-1 2009 The active site electronic structure of the azide complex of substrate-bound human heme oxygenase 1 (hHO) has been investigated by (1)H NMR spectroscopy to shed light on the orbital/spin ground state as an indicator of the unique distal pocket environment of the enzyme. Azides 44-49 heme oxygenase 1 Homo sapiens 83-99 18485456-2 2009 METHODS: Heme oxygenase-1 expression was investigated in the human renal epithelial cell line A498 in response to the uropathogenic Escherichia coli (UPEC) strain IA2, the NO-donor DETA/NONOate (DETA/NO), and proinflammatory cytokines (interleukin-1beta, tumor necrosis factor-alpha, and interferon-gamma) using reverse transcriptase polymerase chain reaction and Western blot analysis. DEET 195-199 heme oxygenase 1 Homo sapiens 9-25 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. Hemin 26-31 heme oxygenase 1 Homo sapiens 13-17 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. Hemin 26-31 heme oxygenase 1 Homo sapiens 54-58 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. Hemin 26-31 heme oxygenase 1 Homo sapiens 54-58 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. DEET 36-40 heme oxygenase 1 Homo sapiens 54-58 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. DEET 36-40 heme oxygenase 1 Homo sapiens 54-58 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. Glutathione 89-100 heme oxygenase 1 Homo sapiens 13-17 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. DEET 160-164 heme oxygenase 1 Homo sapiens 13-17 18485456-4 2009 RESULTS: The HO-1 inducer hemin and DETA/NO increased HO-1 expression in A498 cells, and glutathione depletion further increased HO-1 expression in response to DETA/NO and hemin. Hemin 172-177 heme oxygenase 1 Homo sapiens 13-17 18485456-9 2009 CONCLUSIONS: The expression of HO-1 increased in human renal epithelial cells in response to NO, and the expression was further enhanced in glutathione-depleted cells. Glutathione 140-151 heme oxygenase 1 Homo sapiens 31-35 19389234-1 2009 BACKGROUND: Heme oxygenase-1 is an inducible cytoprotective enzyme which handles oxidative stress by generating anti-oxidant bilirubin and vasodilating carbon monoxide. Bilirubin 125-134 heme oxygenase 1 Homo sapiens 12-28 19389234-1 2009 BACKGROUND: Heme oxygenase-1 is an inducible cytoprotective enzyme which handles oxidative stress by generating anti-oxidant bilirubin and vasodilating carbon monoxide. Carbon Monoxide 152-167 heme oxygenase 1 Homo sapiens 12-28 19389234-2 2009 A (GT)n dinucleotide repeat and a -413A>T single nucleotide polymorphism have been reported in the promoter region of HMOX1 to both influence the occurrence of coronary artery disease and myocardial infarction. (gt)n dinucleotide 2-20 heme oxygenase 1 Homo sapiens 121-126 19239904-0 2009 Sofalcone, an anti-ulcer chalcone derivative, suppresses inflammatory crosstalk between macrophages and adipocytes and adipocyte differentiation: implication of heme-oxygenase-1 induction. sofalcone 0-9 heme oxygenase 1 Homo sapiens 161-177 19239904-4 2009 The suppressive effect of sofalcone on NO production was attenuated by treatment with tin-protoporphyrin (SnPP), a heme-oxygenase (HO)-1 inhibitor. sofalcone 26-35 heme oxygenase 1 Homo sapiens 115-136 19239904-4 2009 The suppressive effect of sofalcone on NO production was attenuated by treatment with tin-protoporphyrin (SnPP), a heme-oxygenase (HO)-1 inhibitor. tin protoporphyrin IX 86-104 heme oxygenase 1 Homo sapiens 115-136 19239904-4 2009 The suppressive effect of sofalcone on NO production was attenuated by treatment with tin-protoporphyrin (SnPP), a heme-oxygenase (HO)-1 inhibitor. S-Nitroso-N-propionyl-D,L-penicillamine 106-110 heme oxygenase 1 Homo sapiens 115-136 19371606-0 2009 Induction of heme oxygenase 1 by arsenite inhibits cytokine-induced monocyte adhesion to human endothelial cells. arsenite 33-41 heme oxygenase 1 Homo sapiens 13-29 19371606-2 2009 Arsenite, as an oxidative stressor, is a potent inducer of HO-1 in human and rodent cells. arsenite 0-8 heme oxygenase 1 Homo sapiens 59-63 19371606-3 2009 In this study, we investigated the mechanistic role of arsenite-induced HO-1 in modulating tumor necrosis factor alpha (TNF-alpha) induced monocyte adhesion to human umbilical vein endothelial cells (HUVEC). arsenite 55-63 heme oxygenase 1 Homo sapiens 72-76 19371606-4 2009 Arsenite pretreatment, which upregulated HO-1 in a time- and concentration-dependent manner, inhibited TNF-alpha-induced monocyte adhesion to HUVEC and intercellular adhesion molecule 1 protein expression by 50% and 40%, respectively. arsenite 0-8 heme oxygenase 1 Homo sapiens 41-45 19371606-5 2009 Importantly, knockdown of HO-1 by small interfering RNA abolished the arsenite-induced inhibitory effects. arsenite 70-78 heme oxygenase 1 Homo sapiens 26-30 19371606-6 2009 These results indicate that induction of HO-1 by arsenite inhibits the cytokine-induced monocyte adhesion to HUVEC by suppressing adhesion molecule expression. arsenite 49-57 heme oxygenase 1 Homo sapiens 41-45 19371606-7 2009 These findings established an important mechanistic link between the functional monocyte adhesion properties of HUVEC and the induction of HO-1 by arsenite. arsenite 147-155 heme oxygenase 1 Homo sapiens 139-143 19336889-1 2009 We previously reported that eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) extracted from Artemisia asiaitica, augmented the cellular antioxidant defense capacity through induction of the antioxidant protein heme oxygenase-1 (HO-1), thereby protecting ileal smooth muscle cells from nonsteroidal anti-inflammatory drug (NSAID)-induced intestinal toxicity. eupatilin 28-37 heme oxygenase 1 Homo sapiens 211-227 19336889-10 2009 We confirmed that ZnPP, an HO-1 inhibitor, repressed eupatilin-induced HO-1 activity and showed the protective effect of eupatilin against indomethacin-induced cell injury. eupatilin 53-62 heme oxygenase 1 Homo sapiens 71-75 19336889-1 2009 We previously reported that eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) extracted from Artemisia asiaitica, augmented the cellular antioxidant defense capacity through induction of the antioxidant protein heme oxygenase-1 (HO-1), thereby protecting ileal smooth muscle cells from nonsteroidal anti-inflammatory drug (NSAID)-induced intestinal toxicity. eupatilin 28-37 heme oxygenase 1 Homo sapiens 229-233 19336889-11 2009 The data suggested that HO-1 was partly responsible for the eupatilin-mediated protective action of esophageal epithelial cells against indomethacin via both ERKs and PI3K/Akt pathways as well as Nrf2 translocation. eupatilin 60-69 heme oxygenase 1 Homo sapiens 24-28 19336889-1 2009 We previously reported that eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) extracted from Artemisia asiaitica, augmented the cellular antioxidant defense capacity through induction of the antioxidant protein heme oxygenase-1 (HO-1), thereby protecting ileal smooth muscle cells from nonsteroidal anti-inflammatory drug (NSAID)-induced intestinal toxicity. eupatilin 39-76 heme oxygenase 1 Homo sapiens 211-227 19336889-11 2009 The data suggested that HO-1 was partly responsible for the eupatilin-mediated protective action of esophageal epithelial cells against indomethacin via both ERKs and PI3K/Akt pathways as well as Nrf2 translocation. Indomethacin 136-148 heme oxygenase 1 Homo sapiens 24-28 19336889-1 2009 We previously reported that eupatilin (5,7-dihydroxy-3,4,6-trimethoxyflavone) extracted from Artemisia asiaitica, augmented the cellular antioxidant defense capacity through induction of the antioxidant protein heme oxygenase-1 (HO-1), thereby protecting ileal smooth muscle cells from nonsteroidal anti-inflammatory drug (NSAID)-induced intestinal toxicity. eupatilin 39-76 heme oxygenase 1 Homo sapiens 229-233 19336889-7 2009 Eupatilin-induced HO-1 expression and Nrf2 were partly attenuated by MEK inhibitor PD98059 and almost completely by phosphatidyl-inactiol 3 kinase (PI3K) inhibitor LY294002, but not by c-Jun N-terminal kinase (JNK) inhibitor SP600125 or p38 mitogen activated protein kinase (MAPK) inhibitor SB202190. eupatilin 0-9 heme oxygenase 1 Homo sapiens 18-22 19336889-7 2009 Eupatilin-induced HO-1 expression and Nrf2 were partly attenuated by MEK inhibitor PD98059 and almost completely by phosphatidyl-inactiol 3 kinase (PI3K) inhibitor LY294002, but not by c-Jun N-terminal kinase (JNK) inhibitor SP600125 or p38 mitogen activated protein kinase (MAPK) inhibitor SB202190. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 83-90 heme oxygenase 1 Homo sapiens 18-22 19336889-7 2009 Eupatilin-induced HO-1 expression and Nrf2 were partly attenuated by MEK inhibitor PD98059 and almost completely by phosphatidyl-inactiol 3 kinase (PI3K) inhibitor LY294002, but not by c-Jun N-terminal kinase (JNK) inhibitor SP600125 or p38 mitogen activated protein kinase (MAPK) inhibitor SB202190. pyrazolanthrone 225-233 heme oxygenase 1 Homo sapiens 18-22 19336889-7 2009 Eupatilin-induced HO-1 expression and Nrf2 were partly attenuated by MEK inhibitor PD98059 and almost completely by phosphatidyl-inactiol 3 kinase (PI3K) inhibitor LY294002, but not by c-Jun N-terminal kinase (JNK) inhibitor SP600125 or p38 mitogen activated protein kinase (MAPK) inhibitor SB202190. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 291-299 heme oxygenase 1 Homo sapiens 18-22 19336889-10 2009 We confirmed that ZnPP, an HO-1 inhibitor, repressed eupatilin-induced HO-1 activity and showed the protective effect of eupatilin against indomethacin-induced cell injury. zinc protoporphyrin 18-22 heme oxygenase 1 Homo sapiens 27-31 19336889-10 2009 We confirmed that ZnPP, an HO-1 inhibitor, repressed eupatilin-induced HO-1 activity and showed the protective effect of eupatilin against indomethacin-induced cell injury. zinc protoporphyrin 18-22 heme oxygenase 1 Homo sapiens 71-75 19336889-10 2009 We confirmed that ZnPP, an HO-1 inhibitor, repressed eupatilin-induced HO-1 activity and showed the protective effect of eupatilin against indomethacin-induced cell injury. eupatilin 53-62 heme oxygenase 1 Homo sapiens 27-31 19407950-9 2009 Compared with the SP600125 group, the expressions of JNK, c-Jun, P-c-Jun and HO-1 mRNA and its protein in the TXLSP+SP600125 group were significantly increased at different time points (P<0.05, P<0.01). pyrazolanthrone 116-124 heme oxygenase 1 Homo sapiens 77-81 19131520-4 2009 This allows measurement of bilirubin formation after incorporation of full-length CPR and heme oxygenase-1 (HO-1) into a membrane environment. Bilirubin 27-36 heme oxygenase 1 Homo sapiens 90-106 19131520-4 2009 This allows measurement of bilirubin formation after incorporation of full-length CPR and heme oxygenase-1 (HO-1) into a membrane environment. Bilirubin 27-36 heme oxygenase 1 Homo sapiens 108-112 19131520-9 2009 These results not only show that HO-1-generated hydrogen peroxide leads to a decrease in HO-1 activity but also provide for a chemically defined system to be used to examine the function of full-length HO-1 in a membrane environment. Hydrogen Peroxide 48-65 heme oxygenase 1 Homo sapiens 33-37 19131520-9 2009 These results not only show that HO-1-generated hydrogen peroxide leads to a decrease in HO-1 activity but also provide for a chemically defined system to be used to examine the function of full-length HO-1 in a membrane environment. Hydrogen Peroxide 48-65 heme oxygenase 1 Homo sapiens 89-93 19131520-9 2009 These results not only show that HO-1-generated hydrogen peroxide leads to a decrease in HO-1 activity but also provide for a chemically defined system to be used to examine the function of full-length HO-1 in a membrane environment. Hydrogen Peroxide 48-65 heme oxygenase 1 Homo sapiens 89-93 19243019-8 2009 Among polymorphisms in other genes, only the (GT)n repeat polymorphism in the HMOX1 promoter region showed association with TBIL levels in the Uyghur population, but not in the Han and Kazak populations. Bilirubin 124-128 heme oxygenase 1 Homo sapiens 78-83 19136476-0 2009 15-Deoxy-Delta12,14-prostaglandin J2 upregulates the expression of heme oxygenase-1 and subsequently matrix metalloproteinase-1 in human breast cancer cells: possible roles of iron and ROS. Iron 176-180 heme oxygenase 1 Homo sapiens 67-127 19136476-0 2009 15-Deoxy-Delta12,14-prostaglandin J2 upregulates the expression of heme oxygenase-1 and subsequently matrix metalloproteinase-1 in human breast cancer cells: possible roles of iron and ROS. Reactive Oxygen Species 185-188 heme oxygenase 1 Homo sapiens 67-127 19136476-3 2009 Treatment of MCF-7 and MDA-MB-231 cells with 30 microM of 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2) increased the expression of HO-1, which preceded the induction of matrix metalloproteinases (MMPs). 14-prostaglandin j2 75-94 heme oxygenase 1 Homo sapiens 134-138 19136476-4 2009 The 15d-PGJ2-induced upregulation of MMP-1 was abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) as well as introduction of HO-1 short interfering RNA. 15-deoxy-delta(12,14)-prostaglandin J2 4-12 heme oxygenase 1 Homo sapiens 64-68 19136476-4 2009 The 15d-PGJ2-induced upregulation of MMP-1 was abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) as well as introduction of HO-1 short interfering RNA. 15-deoxy-delta(12,14)-prostaglandin J2 4-12 heme oxygenase 1 Homo sapiens 136-140 19136476-4 2009 The 15d-PGJ2-induced upregulation of MMP-1 was abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) as well as introduction of HO-1 short interfering RNA. zinc protoporphyrin 79-101 heme oxygenase 1 Homo sapiens 64-68 19136476-4 2009 The 15d-PGJ2-induced upregulation of MMP-1 was abrogated by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP) as well as introduction of HO-1 short interfering RNA. zinc protoporphyrin 103-107 heme oxygenase 1 Homo sapiens 64-68 19136476-5 2009 In addition, HO-1 inducers, such as cobalt protoporphyrin IX and hemin, upregulated the expression of MMP-1. cobaltiprotoporphyrin 36-60 heme oxygenase 1 Homo sapiens 13-17 19136476-7 2009 Treatment with the HO-1 inhibitor ZnPP abolished the migrative phenotype of 15d-PGJ2-treated MCF-7 cells. zinc protoporphyrin 34-38 heme oxygenase 1 Homo sapiens 19-23 19136476-7 2009 Treatment with the HO-1 inhibitor ZnPP abolished the migrative phenotype of 15d-PGJ2-treated MCF-7 cells. 9-deoxy-delta-9-prostaglandin D2 80-84 heme oxygenase 1 Homo sapiens 19-23 19136476-9 2009 We hypothesize that excess iron, released as a consequence HO-1 activity induced by 15d-PGJ2, is transiently available for the stimulation of intracellular ROS generation and subsequently MMP-1 expression. Iron 27-31 heme oxygenase 1 Homo sapiens 59-63 19136476-9 2009 We hypothesize that excess iron, released as a consequence HO-1 activity induced by 15d-PGJ2, is transiently available for the stimulation of intracellular ROS generation and subsequently MMP-1 expression. 15-deoxy-delta(12,14)-prostaglandin J2 84-92 heme oxygenase 1 Homo sapiens 59-63 19136476-9 2009 We hypothesize that excess iron, released as a consequence HO-1 activity induced by 15d-PGJ2, is transiently available for the stimulation of intracellular ROS generation and subsequently MMP-1 expression. Reactive Oxygen Species 156-159 heme oxygenase 1 Homo sapiens 59-63 19136476-12 2009 In conclusion, 15d-PGJ2 upregulates MMP-1 expression via induction of HO-1 and subsequent production of iron capable of generating ROS, which may contribute to increased metastasis and invasiveness of the human breast cancer cells. 15-deoxy-delta(12,14)-prostaglandin J2 15-23 heme oxygenase 1 Homo sapiens 70-74 18952926-0 2009 Reactivation of optic nerve head astrocytes by TGF-beta2 and H2O2 is accompanied by increased Hsp32 and Hsp47 expression. Hydrogen Peroxide 61-65 heme oxygenase 1 Homo sapiens 94-99 18952926-8 2009 Exposure of the cells to H(2)O(2) could increase both Hsp32 and -47. Hydrogen Peroxide 25-33 heme oxygenase 1 Homo sapiens 54-59 18996220-0 2009 Sodium arsenite induces ROS generation, DNA oxidative damage, HO-1 and c-Myc proteins, NF-kappaB activation and cell proliferation in human breast cancer MCF-7 cells. sodium arsenite 0-15 heme oxygenase 1 Homo sapiens 62-66 19238116-2 2009 This study investigated the genetic variants of four bilirubin metabolism genes--heme oxygenase-1 (HMOX1), biliverdin reductase A (BLVRA), solute carrier organic anion transporter family member 1B1 (SLCO1B1), and uridine diphosphate glycosyltransferase 1A1 (UGT1A1)--in relation to TBIL levels and CAD. Bilirubin 53-62 heme oxygenase 1 Homo sapiens 99-104 18996220-5 2009 ROS-mediated DNA damage as measured by the presence of 8-OHdG DNA-adducts in their nuclei, IkappaB phosphorylation, NF-kappaB activation and increases in c-Myc and HO-1 protein levels were also observed, suggesting that these factors play a relevant role in the arsenite induced MCF-7 cell recruitment into the S-phase of the cell cycle and cell proliferation observed. ros 0-3 heme oxygenase 1 Homo sapiens 164-168 19374863-6 2009 Together with AG-126, structurally related benzylidenemalononitrile tyrphostins AG-9, AG-10, AG-18, and AG-1288 were able to up-regulate the expression of HO-1 and several other genes, although with relatively less efficacy. Benzylidenemalononitrile 43-67 heme oxygenase 1 Homo sapiens 155-159 19174156-0 2009 KB-34, a newly synthesized chalcone derivative, inhibits lipopolysaccharide-stimulated nitric oxide production in RAW 264.7 macrophages via heme oxygenase-1 induction and blockade of activator protein-1. kb-34 0-5 heme oxygenase 1 Homo sapiens 140-156 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. S-Nitroso-N-propionyl-D,L-penicillamine 15-19 heme oxygenase 1 Homo sapiens 46-50 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. S-Nitroso-N-propionyl-D,L-penicillamine 15-19 heme oxygenase 1 Homo sapiens 130-134 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. kb-34 65-70 heme oxygenase 1 Homo sapiens 46-50 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. kb-34 65-70 heme oxygenase 1 Homo sapiens 130-134 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. Nitrites 94-101 heme oxygenase 1 Homo sapiens 46-50 19174156-7 2009 Treatment with SnPP, a selective inhibitor of HO-1, reversed the KB-34-mediated inhibition of nitrite production, suggesting that HO-1 plays an important role in the suppression of NO production by KB-34. Nitrites 94-101 heme oxygenase 1 Homo sapiens 130-134 19154785-8 2009 UFP also up-regulated heme oxygenase-1 (HO-1) and tissue factor (TF) mRNA expression, and pretreatment with the antioxidant N-acetylcysteine significantly decreased their expression. Acetylcysteine 124-140 heme oxygenase 1 Homo sapiens 22-38 19154785-10 2009 Treatment with the JNK inhibitor SP600125 and silencing of both JNK1 and JNK2 with siRNA inhibited UFP-stimulated O(2)(-) production and mRNA expression of HO-1 and TF. pyrazolanthrone 33-41 heme oxygenase 1 Homo sapiens 156-167 19374863-6 2009 Together with AG-126, structurally related benzylidenemalononitrile tyrphostins AG-9, AG-10, AG-18, and AG-1288 were able to up-regulate the expression of HO-1 and several other genes, although with relatively less efficacy. Tyrphostins 68-79 heme oxygenase 1 Homo sapiens 155-159 19135260-0 2009 Covalent heme attachment to the protein in human heme oxygenase-1 with selenocysteine replacing the His25 proximal iron ligand. Heme 9-13 heme oxygenase 1 Homo sapiens 49-65 19100829-7 2009 OxLDL-mediated induction of the oxidative stress responsive gene, heme oxygenase-1, was also abolished by H(2)S. Hydrogen Sulfide 106-111 heme oxygenase 1 Homo sapiens 66-82 19299217-5 2009 According to our previous study, AVF failure was associated with a longer dinucleotide (GT)n repeat (n > or = 30) in the promoter of the heme oxygenase-1 (HO-1) gene. Dinucleoside Phosphates 74-86 heme oxygenase 1 Homo sapiens 140-156 19299217-5 2009 According to our previous study, AVF failure was associated with a longer dinucleotide (GT)n repeat (n > or = 30) in the promoter of the heme oxygenase-1 (HO-1) gene. Dinucleoside Phosphates 74-86 heme oxygenase 1 Homo sapiens 158-162 19236154-3 2009 Data generated by various groups indicate that Nrf2 induces the expression of a group of cytoprotective, antixenobiotic and antioxidant enzymes that include heme oxygenase-1, NAD(P)H:quinone oxidoreductase and enzymes of glutathione (GSH) metabolism such as gamma-glutamyl cysteine ligase, GSH transferases and so on. Glutathione 221-232 heme oxygenase 1 Homo sapiens 157-173 19236154-3 2009 Data generated by various groups indicate that Nrf2 induces the expression of a group of cytoprotective, antixenobiotic and antioxidant enzymes that include heme oxygenase-1, NAD(P)H:quinone oxidoreductase and enzymes of glutathione (GSH) metabolism such as gamma-glutamyl cysteine ligase, GSH transferases and so on. Glutathione 234-237 heme oxygenase 1 Homo sapiens 157-173 19212657-1 2009 Human heme oxygenase-1 (hHO-1) is a rate-limiting enzyme in heme metabolism. Heme 6-10 heme oxygenase 1 Homo sapiens 24-29 19212657-3 2009 Site-directed mutagenesis studies indicate that the proximal residue histidine 25 (His25) plays a key role in hHO-1 activity. Histidine 69-78 heme oxygenase 1 Homo sapiens 110-115 19212657-10 2009 By supplementing imidazole, the HO-1 activity was restored approximately 90% to its normal level. imidazole 17-26 heme oxygenase 1 Homo sapiens 32-36 19135260-0 2009 Covalent heme attachment to the protein in human heme oxygenase-1 with selenocysteine replacing the His25 proximal iron ligand. Selenocysteine 71-85 heme oxygenase 1 Homo sapiens 49-65 19135260-0 2009 Covalent heme attachment to the protein in human heme oxygenase-1 with selenocysteine replacing the His25 proximal iron ligand. Iron 115-119 heme oxygenase 1 Homo sapiens 49-65 19135260-1 2009 To characterize heme oxygenase with a selenocysteine (SeCys) as the proximal iron ligand, we have expressed truncated human heme oxygenase-1 (hHO-1) His25Cys, in which Cys-25 is the only cysteine, in the Escherichia coli cysteine auxotroph strain BL21(DE3)cys. Selenocysteine 38-52 heme oxygenase 1 Homo sapiens 124-140 19135260-1 2009 To characterize heme oxygenase with a selenocysteine (SeCys) as the proximal iron ligand, we have expressed truncated human heme oxygenase-1 (hHO-1) His25Cys, in which Cys-25 is the only cysteine, in the Escherichia coli cysteine auxotroph strain BL21(DE3)cys. Selenocysteine 38-52 heme oxygenase 1 Homo sapiens 142-147 19135260-5 2009 The heme-His25SeCys hHO-1 complex could be prepared by either (a) supplementing the overexpression medium with heme, or (b) reconstituting the purified apoprotein with heme. Heme 4-8 heme oxygenase 1 Homo sapiens 20-25 19135260-5 2009 The heme-His25SeCys hHO-1 complex could be prepared by either (a) supplementing the overexpression medium with heme, or (b) reconstituting the purified apoprotein with heme. Heme 111-115 heme oxygenase 1 Homo sapiens 20-25 19135260-11 2009 47 (2008) 3480-3482 ], indicate that a selenyl radical is formed in the hHO-1 His25SeCys mutant that adds to a heme vinyl group. selenyl radical 39-54 heme oxygenase 1 Homo sapiens 72-77 19135260-11 2009 47 (2008) 3480-3482 ], indicate that a selenyl radical is formed in the hHO-1 His25SeCys mutant that adds to a heme vinyl group. Heme 111-115 heme oxygenase 1 Homo sapiens 72-77 19122175-0 2009 Induction of prostacyclin by steady laminar shear stress suppresses tumor necrosis factor-alpha biosynthesis via heme oxygenase-1 in human endothelial cells. Epoprostenol 13-25 heme oxygenase 1 Homo sapiens 113-129 19129475-5 2009 Although the first phase of HO-1 induction at 6 h was accomplished by AC-derived sphingosine-1-phosphate (S1P) acting via S1P receptor 1, the second wave of HO-1 induction at 24 h was attributed to autocrine signaling of vascular endothelial growth factor A (VEGFA), whose expression and release were facilitated by S1P. Charcoal 70-72 heme oxygenase 1 Homo sapiens 28-32 19129475-5 2009 Although the first phase of HO-1 induction at 6 h was accomplished by AC-derived sphingosine-1-phosphate (S1P) acting via S1P receptor 1, the second wave of HO-1 induction at 24 h was attributed to autocrine signaling of vascular endothelial growth factor A (VEGFA), whose expression and release were facilitated by S1P. sphingosine 1-phosphate 81-104 heme oxygenase 1 Homo sapiens 28-32 19417726-3 2009 It has been clinically demonstrated that the immunoreactivities of intrarenal heme oxygenase-1 (HO-1) and 4-hydroxy-2-nonenal (4-HNE) markers of reactive oxygen species (ROS) and those of intrarenal angiotensinogen (AGT) and angiotensin II (Ang II) markers of renin angiotensin system (RAS) in IgA nephropathy patients were significantly increased as compared to those of control subjects. Reactive Oxygen Species 145-168 heme oxygenase 1 Homo sapiens 78-94 19417726-3 2009 It has been clinically demonstrated that the immunoreactivities of intrarenal heme oxygenase-1 (HO-1) and 4-hydroxy-2-nonenal (4-HNE) markers of reactive oxygen species (ROS) and those of intrarenal angiotensinogen (AGT) and angiotensin II (Ang II) markers of renin angiotensin system (RAS) in IgA nephropathy patients were significantly increased as compared to those of control subjects. Reactive Oxygen Species 170-173 heme oxygenase 1 Homo sapiens 78-94 19122175-8 2009 Carbacyclin, an agonist of IP, induced HO-1. carboprostacyclin 0-11 heme oxygenase 1 Homo sapiens 39-43 19122175-9 2009 Similarly to inhibition of prostacyclin biosynthesis or activity, the novel imidazole-based HO-1 inhibitor QC15 reversed TNF-alpha reduction by LSS. imidazole 76-85 heme oxygenase 1 Homo sapiens 92-96 19122175-10 2009 These findings suggest that inhibition of COX-2-dependent prostacyclin might contribute to acceleration of atherogenesis in patients taking traditional nonsteroidal antiinflammatory drugs (NSAIDs) and NSAIDs selective for COX-2 through downregulation of HO-1, which halts TNF-alpha generation in human endothelial cells. Epoprostenol 58-70 heme oxygenase 1 Homo sapiens 254-258 19033392-9 2009 A significant inhibition of the expression of hmox1 and nqo1 mRNAs and CD86 expression was found in 1-chloro 2,4-dinitrobenzene-treated THP-1 cells preincubated with N-acetyl cysteine, a glutathione precursor. Dinitrochlorobenzene 100-127 heme oxygenase 1 Homo sapiens 46-51 18842672-11 2009 CONCLUSIONS: Collectively, we suggest that HO-1 induced by TMP might, at least in part, protect against gentamicin-induced nephrotoxicity through antiapoptotic and anti-inflammatory mechanisms, and that it may have therapeutic potential for patients with renal disease. tetramethylpyrazine 59-62 heme oxygenase 1 Homo sapiens 43-47 18842672-11 2009 CONCLUSIONS: Collectively, we suggest that HO-1 induced by TMP might, at least in part, protect against gentamicin-induced nephrotoxicity through antiapoptotic and anti-inflammatory mechanisms, and that it may have therapeutic potential for patients with renal disease. Gentamicins 104-114 heme oxygenase 1 Homo sapiens 43-47 19036700-0 2009 Heme oxygenase-1 expression enhances vascular endothelial resistance to complement-mediated injury through induction of decay-accelerating factor: a role for increased bilirubin and ferritin. Bilirubin 168-177 heme oxygenase 1 Homo sapiens 0-16 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Heme 19-23 heme oxygenase 1 Homo sapiens 27-43 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Heme 19-23 heme oxygenase 1 Homo sapiens 45-49 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 27-43 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 45-49 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Biliverdine 78-88 heme oxygenase 1 Homo sapiens 27-43 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Biliverdine 78-88 heme oxygenase 1 Homo sapiens 45-49 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Iron 99-103 heme oxygenase 1 Homo sapiens 27-43 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Iron 99-103 heme oxygenase 1 Homo sapiens 45-49 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Iron 105-107 heme oxygenase 1 Homo sapiens 27-43 19036700-1 2009 Catabolism of free heme by heme oxygenase-1 (HO-1) generates carbon monoxide, biliverdin, and free iron (Fe). Iron 105-107 heme oxygenase 1 Homo sapiens 45-49 19036700-5 2009 In contrast, HO-1 agonists hemin and cobalt protoporphyrin IX significantly increased DAF protein expression, reflecting an increase in transcription and steady-state mRNA. Cobalt 37-43 heme oxygenase 1 Homo sapiens 13-17 19036700-5 2009 In contrast, HO-1 agonists hemin and cobalt protoporphyrin IX significantly increased DAF protein expression, reflecting an increase in transcription and steady-state mRNA. protoporphyrin IX 44-61 heme oxygenase 1 Homo sapiens 13-17 19036700-6 2009 Adenoviral-mediated overexpression of HO-1 increased DAF expression, enhancing protection against C3 deposition and complement-mediated lysis, and this was reversed by DAF inhibitory monoclonal antibody (mAb) 1H4. 1H4 209-212 heme oxygenase 1 Homo sapiens 38-42 18845641-6 2009 Real-time PCR analyses, Western blotting, and immunofluorescence of first trimester placentae and differentiating villous explant cultures demonstrated down-regulation of HO-1 in invasive EVTs as compared with CTBs. evts 188-192 heme oxygenase 1 Homo sapiens 171-175 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Heme 130-134 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Iron 140-152 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Carbon Monoxide 154-169 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Carbon Monoxide 171-173 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Biliverdine 180-190 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Biliverdine 192-194 heme oxygenase 1 Homo sapiens 0-21 20107533-1 2009 Heme oxygenase (HO)-1 is an inducible enzyme that catalyzes the first and rate-limiting step in the oxidative degradation of free heme into ferrous iron, carbon monoxide (CO), and biliverdin (BV), the latter being subsequently converted into bilirubin (BR). Bilirubin 242-251 heme oxygenase 1 Homo sapiens 0-21 18757839-6 2009 We hypothesized that PMET activation by Ox-PAPC causes intracellular NAD(P)H depletion, which leads to the increased oxidative stress and HO-1 induction. nad(p)h 69-76 heme oxygenase 1 Homo sapiens 138-142 18757839-7 2009 Supporting this hypothesis, cotreatment of cells with exogenous NAD(P)H and Ox-PAPC significantly decreased oxidative stress and HO-1 induction by Ox-PAPC. nad(p)h 64-71 heme oxygenase 1 Homo sapiens 129-133 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Heme 76-80 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Heme 76-80 heme oxygenase 1 Homo sapiens 18-22 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 18-22 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Iron 103-107 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Iron 103-107 heme oxygenase 1 Homo sapiens 18-22 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Iron 166-170 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Iron 166-170 heme oxygenase 1 Homo sapiens 18-22 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Biliverdine 197-207 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Biliverdine 197-207 heme oxygenase 1 Homo sapiens 18-22 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Bilirubin 231-240 heme oxygenase 1 Homo sapiens 0-16 19162549-1 2009 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that catabolizes free heme into carbon monoxide, iron (which induces the expression of heavy-chain ferritin, an iron-sequestering protein) and biliverdin (which is converted to bilirubin by biliverdin reductase). Bilirubin 231-240 heme oxygenase 1 Homo sapiens 18-22 19162549-2 2009 Over the past few years it has become apparent that these "arms" of the HO-1 system can act protectively in a variety of experimental models of disease; there is also evidence that HO-1 and bilirubin have protective actions in humans. Bilirubin 190-199 heme oxygenase 1 Homo sapiens 72-76 19162549-3 2009 Here, we present a model for the beneficial actions of the products of heme degradation, and we discuss the potential clinical applications of enhancing the HO-1 system. Heme 71-75 heme oxygenase 1 Homo sapiens 157-161 18812229-7 2009 Both inducible nitric oxide synthase (NOS-2) and hemoxygenase-1 (HO-1) gene expression were increased by tBH and reduced by both RES and SM pretreatments. tert-Butylhydroperoxide 105-108 heme oxygenase 1 Homo sapiens 49-69 19033392-9 2009 A significant inhibition of the expression of hmox1 and nqo1 mRNAs and CD86 expression was found in 1-chloro 2,4-dinitrobenzene-treated THP-1 cells preincubated with N-acetyl cysteine, a glutathione precursor. Acetylcysteine 166-183 heme oxygenase 1 Homo sapiens 46-51 19033392-9 2009 A significant inhibition of the expression of hmox1 and nqo1 mRNAs and CD86 expression was found in 1-chloro 2,4-dinitrobenzene-treated THP-1 cells preincubated with N-acetyl cysteine, a glutathione precursor. Glutathione 187-198 heme oxygenase 1 Homo sapiens 46-51 18996444-7 2009 ODQ and KT5823 decreased, whereas 8-bromo-cyclic GMP increased, the number of dopaminergic neurons expressing HO-1 after IFN-gamma/LPS challenge, without parallel changes in HO-1 expression in other cell populations. 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one 0-3 heme oxygenase 1 Homo sapiens 110-114 18996444-0 2009 Nitric oxide-cyclic GMP signaling pathway limits inflammatory degeneration of midbrain dopaminergic neurons: cell type-specific regulation of heme oxygenase-1 expression. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 142-158 18996444-8 2009 An NO donor 3-(4-morpholinyl)sydnonimine hydrochloride also induced HO-1 expression in dopaminergic neurons, which was abolished by ODQ and augmented by 8-bromo-cyclic GMP. 3-(4-morpholinyl)sydnonimine hydrochloride 12-54 heme oxygenase 1 Homo sapiens 68-72 18996444-8 2009 An NO donor 3-(4-morpholinyl)sydnonimine hydrochloride also induced HO-1 expression in dopaminergic neurons, which was abolished by ODQ and augmented by 8-bromo-cyclic GMP. 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one 132-135 heme oxygenase 1 Homo sapiens 68-72 18996444-9 2009 Moreover, IFN-gamma/LPS-induced dopaminergic cell death was augmented by zinc protoporphyrin IX, an HO-1 inhibitor. zinc protoporphyrin 73-95 heme oxygenase 1 Homo sapiens 100-104 18845130-0 2009 Reactive oxygen species-independent apoptosis in doxorubicin-treated H9c2 cardiomyocytes: role for heme oxygenase-1 down-modulation. Doxorubicin 49-60 heme oxygenase 1 Homo sapiens 99-115 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Nitric Oxide 36-48 heme oxygenase 1 Homo sapiens 150-166 18992762-5 2009 CDDP significantly increased renal abundances of HO-1, HSP60, HSP72 and HSP90 at days 1, 3, and 5. Cisplatin 0-4 heme oxygenase 1 Homo sapiens 49-53 19092646-6 2009 Both HO-1 and ferritin have been identified as targets of, and inducible by, aspirin and, in the case of HO-1, aspirin-triggered lipoxins. Aspirin 77-84 heme oxygenase 1 Homo sapiens 5-9 19092646-6 2009 Both HO-1 and ferritin have been identified as targets of, and inducible by, aspirin and, in the case of HO-1, aspirin-triggered lipoxins. Aspirin 111-118 heme oxygenase 1 Homo sapiens 5-9 19092646-6 2009 Both HO-1 and ferritin have been identified as targets of, and inducible by, aspirin and, in the case of HO-1, aspirin-triggered lipoxins. Aspirin 111-118 heme oxygenase 1 Homo sapiens 105-109 19092646-6 2009 Both HO-1 and ferritin have been identified as targets of, and inducible by, aspirin and, in the case of HO-1, aspirin-triggered lipoxins. Lipoxins 129-137 heme oxygenase 1 Homo sapiens 5-9 19092646-6 2009 Both HO-1 and ferritin have been identified as targets of, and inducible by, aspirin and, in the case of HO-1, aspirin-triggered lipoxins. Lipoxins 129-137 heme oxygenase 1 Homo sapiens 105-109 19092646-8 2009 HO-1 and its antioxidant product bilirubin have been reported to be not only involved in vasoprotection, but to have a similar function in gastric tissue. Bilirubin 33-42 heme oxygenase 1 Homo sapiens 0-4 19183882-0 2009 NS398 protects cells from sodium nitroprusside-mediated cytotoxicity through enhancing HO-1 induction independent of COX-2 inhibition. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 0-5 heme oxygenase 1 Homo sapiens 87-91 19183882-0 2009 NS398 protects cells from sodium nitroprusside-mediated cytotoxicity through enhancing HO-1 induction independent of COX-2 inhibition. Nitroprusside 26-46 heme oxygenase 1 Homo sapiens 87-91 19183882-3 2009 Recently, however, salicylate the active metabolite of aspirin showed COX-independent anti-inflammatory effects through induction of HO-1. Salicylates 19-29 heme oxygenase 1 Homo sapiens 133-137 19183882-3 2009 Recently, however, salicylate the active metabolite of aspirin showed COX-independent anti-inflammatory effects through induction of HO-1. Aspirin 55-62 heme oxygenase 1 Homo sapiens 133-137 19183882-4 2009 Thus, we hypothesized that HO-1 are induced as an adaptive response to sodium nitroprusside (SNP) and play a protective role against cytotoxicity. Nitroprusside 71-91 heme oxygenase 1 Homo sapiens 27-31 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Heme 63-67 heme oxygenase 1 Homo sapiens 0-16 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Heme 63-67 heme oxygenase 1 Homo sapiens 18-22 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Heme 63-67 heme oxygenase 1 Homo sapiens 157-161 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Biliverdine 71-81 heme oxygenase 1 Homo sapiens 0-16 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Biliverdine 71-81 heme oxygenase 1 Homo sapiens 18-22 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Biliverdine 71-81 heme oxygenase 1 Homo sapiens 157-161 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 0-16 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 18-22 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 157-161 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Iron 109-113 heme oxygenase 1 Homo sapiens 0-16 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Iron 109-113 heme oxygenase 1 Homo sapiens 18-22 19123922-1 2009 Heme oxygenase-1 (HO-1) catalyzes the oxidative degradation of heme to biliverdin, carbon monoxide, and free iron in a reaction requiring the interaction of HO-1 with NADPH-cytochrome P450 reductase (CPR). Iron 109-113 heme oxygenase 1 Homo sapiens 157-161 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Nitric Oxide 36-48 heme oxygenase 1 Homo sapiens 168-172 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Carbon Monoxide 58-73 heme oxygenase 1 Homo sapiens 150-166 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Carbon Monoxide 58-73 heme oxygenase 1 Homo sapiens 168-172 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Carbon Monoxide 75-77 heme oxygenase 1 Homo sapiens 150-166 19292935-4 2009 It is now generally recognized that nitric oxide (NO) and carbon monoxide (CO), two gaso-transmitters synthesized by inducible NO synthase (iNOS) and heme-oxygenase-1 (HO-1) respectively, play important roles in the compensatory regulation of the blood pressure during the development of hypertension. Carbon Monoxide 75-77 heme oxygenase 1 Homo sapiens 168-172 19888908-5 2009 Exposure of HepG2 and Hep3B cells to Cu2+ inhibited the enzymes PBGD and ALAD of the heme synthesis pathway and, in parallel, upregulated heme oxygenase-1 (HO-1). cupric ion 37-41 heme oxygenase 1 Homo sapiens 138-154 19794825-6 2009 In addition, LAS0811 induced expression of heme oxygenase 1 (HO1), which is an ARE-regulated enzyme with anti-inflammatory activity. las0811 13-20 heme oxygenase 1 Homo sapiens 43-59 19794825-6 2009 In addition, LAS0811 induced expression of heme oxygenase 1 (HO1), which is an ARE-regulated enzyme with anti-inflammatory activity. las0811 13-20 heme oxygenase 1 Homo sapiens 61-64 19177192-0 2009 Dimethoxycurcumin, a Synthetic Curcumin Analogue, Induces Heme Oxygenase-1 Expression through Nrf2 Activation in RAW264.7 Macrophages. dimethoxycurcumin 0-17 heme oxygenase 1 Homo sapiens 58-74 19177192-0 2009 Dimethoxycurcumin, a Synthetic Curcumin Analogue, Induces Heme Oxygenase-1 Expression through Nrf2 Activation in RAW264.7 Macrophages. Curcumin 31-39 heme oxygenase 1 Homo sapiens 58-74 19177192-2 2009 The aim of this study was to investigate whether dimethoxycurcumin [1,7-bis(4,3-dimethoxyphenyl)-1,6-heptadiene-3,5-dione], a synthetic curcumin analogue with higher metabolic stability over curcumin, could induce HO-1 expression to the same extent as curcumin in RAW264.7 macrophages. dimethoxycurcumin 49-66 heme oxygenase 1 Homo sapiens 214-218 19177192-2 2009 The aim of this study was to investigate whether dimethoxycurcumin [1,7-bis(4,3-dimethoxyphenyl)-1,6-heptadiene-3,5-dione], a synthetic curcumin analogue with higher metabolic stability over curcumin, could induce HO-1 expression to the same extent as curcumin in RAW264.7 macrophages. 1,7-bis(4,3-dimethoxyphenyl)-1,6-heptadiene-3,5-dione 68-121 heme oxygenase 1 Homo sapiens 214-218 19177192-2 2009 The aim of this study was to investigate whether dimethoxycurcumin [1,7-bis(4,3-dimethoxyphenyl)-1,6-heptadiene-3,5-dione], a synthetic curcumin analogue with higher metabolic stability over curcumin, could induce HO-1 expression to the same extent as curcumin in RAW264.7 macrophages. Curcumin 58-66 heme oxygenase 1 Homo sapiens 214-218 19177192-3 2009 Dimethoxycurcumin and curcumin, but not tetrahydrocurcumin, induced HO-1 expression and Nrf2 nuclear translocation, suggesting that the unsaturated nature of the diarylheptanoid chain of the compounds are crucial for HO-1 expression and Nrf2 activation. dimethoxycurcumin 0-17 heme oxygenase 1 Homo sapiens 68-72 19177192-3 2009 Dimethoxycurcumin and curcumin, but not tetrahydrocurcumin, induced HO-1 expression and Nrf2 nuclear translocation, suggesting that the unsaturated nature of the diarylheptanoid chain of the compounds are crucial for HO-1 expression and Nrf2 activation. dimethoxycurcumin 0-17 heme oxygenase 1 Homo sapiens 217-221 19177192-3 2009 Dimethoxycurcumin and curcumin, but not tetrahydrocurcumin, induced HO-1 expression and Nrf2 nuclear translocation, suggesting that the unsaturated nature of the diarylheptanoid chain of the compounds are crucial for HO-1 expression and Nrf2 activation. Curcumin 9-17 heme oxygenase 1 Homo sapiens 68-72 19177192-3 2009 Dimethoxycurcumin and curcumin, but not tetrahydrocurcumin, induced HO-1 expression and Nrf2 nuclear translocation, suggesting that the unsaturated nature of the diarylheptanoid chain of the compounds are crucial for HO-1 expression and Nrf2 activation. Curcumin 9-17 heme oxygenase 1 Homo sapiens 217-221 19177192-4 2009 Blockage of Nrf2 synthesis by small interfering RNA abolished HO-1 expression by dimethoxycurcumin, indicating that dimethoxycurcumin may induce HO-1 expression via Nrf2 activation. dimethoxycurcumin 81-98 heme oxygenase 1 Homo sapiens 62-66 19177192-4 2009 Blockage of Nrf2 synthesis by small interfering RNA abolished HO-1 expression by dimethoxycurcumin, indicating that dimethoxycurcumin may induce HO-1 expression via Nrf2 activation. dimethoxycurcumin 81-98 heme oxygenase 1 Homo sapiens 145-149 19177192-4 2009 Blockage of Nrf2 synthesis by small interfering RNA abolished HO-1 expression by dimethoxycurcumin, indicating that dimethoxycurcumin may induce HO-1 expression via Nrf2 activation. dimethoxycurcumin 116-133 heme oxygenase 1 Homo sapiens 62-66 19177192-4 2009 Blockage of Nrf2 synthesis by small interfering RNA abolished HO-1 expression by dimethoxycurcumin, indicating that dimethoxycurcumin may induce HO-1 expression via Nrf2 activation. dimethoxycurcumin 116-133 heme oxygenase 1 Homo sapiens 145-149 19177192-5 2009 In comparison, dimethoxycurcumin and curcumin had about the same effect on HO-1 expression, suggesting that dimethoxycurcumin retains the HO-1-inducing activity of its parent compound curcumin in RAW264.7 macrophages. dimethoxycurcumin 15-32 heme oxygenase 1 Homo sapiens 75-79 19177192-5 2009 In comparison, dimethoxycurcumin and curcumin had about the same effect on HO-1 expression, suggesting that dimethoxycurcumin retains the HO-1-inducing activity of its parent compound curcumin in RAW264.7 macrophages. Curcumin 24-32 heme oxygenase 1 Homo sapiens 75-79 19177192-5 2009 In comparison, dimethoxycurcumin and curcumin had about the same effect on HO-1 expression, suggesting that dimethoxycurcumin retains the HO-1-inducing activity of its parent compound curcumin in RAW264.7 macrophages. dimethoxycurcumin 108-125 heme oxygenase 1 Homo sapiens 75-79 19177192-5 2009 In comparison, dimethoxycurcumin and curcumin had about the same effect on HO-1 expression, suggesting that dimethoxycurcumin retains the HO-1-inducing activity of its parent compound curcumin in RAW264.7 macrophages. dimethoxycurcumin 108-125 heme oxygenase 1 Homo sapiens 138-142 19177192-5 2009 In comparison, dimethoxycurcumin and curcumin had about the same effect on HO-1 expression, suggesting that dimethoxycurcumin retains the HO-1-inducing activity of its parent compound curcumin in RAW264.7 macrophages. Curcumin 37-45 heme oxygenase 1 Homo sapiens 75-79 19355962-6 2009 The anti-inflammatory activity of chalcones has been correlated with the induction of heme oxygenase-1 while phlorotannins have been found to inhibit matrix metalloproteinase, which is implicated in arthritis, chronic inflammation, and wrinkle formation. Chalcones 34-43 heme oxygenase 1 Homo sapiens 86-102 19263288-1 2009 Heme oxygenases (HO-1 and HO-2) are responsible for the production of carbon monoxide, a vasodilator. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 0-30 19177185-3 2009 Cells counteract this by rapidly inducing the rate-limiting enzyme in heme breakdown, heme oxygenase-1 (HO-1), which is a low-molecular-weight stress protein. Heme 70-74 heme oxygenase 1 Homo sapiens 86-102 19177185-3 2009 Cells counteract this by rapidly inducing the rate-limiting enzyme in heme breakdown, heme oxygenase-1 (HO-1), which is a low-molecular-weight stress protein. Heme 70-74 heme oxygenase 1 Homo sapiens 104-108 19177185-4 2009 The enzymatic HO-1 reaction removes heme. Heme 36-40 heme oxygenase 1 Homo sapiens 14-18 19888908-5 2009 Exposure of HepG2 and Hep3B cells to Cu2+ inhibited the enzymes PBGD and ALAD of the heme synthesis pathway and, in parallel, upregulated heme oxygenase-1 (HO-1). cupric ion 37-41 heme oxygenase 1 Homo sapiens 156-160 21152244-9 2009 VEGF, MMP9 and HO-1 transcript levels were increased in ARPE19 cells under H(2)O(2) - induced oxidative stress. Hydrogen Peroxide 75-83 heme oxygenase 1 Homo sapiens 15-19 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. Iron 91-95 heme oxygenase 1 Homo sapiens 51-55 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. ammonium ferrous sulfate 97-103 heme oxygenase 1 Homo sapiens 51-55 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. Carbon Monoxide 106-121 heme oxygenase 1 Homo sapiens 51-55 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. Carbon Monoxide 123-125 heme oxygenase 1 Homo sapiens 51-55 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 51-55 19046352-3 2009 In the current study, we determined the effects of HO-1 over-expression and its byproducts iron (Fe(2+)), carbon monoxide (CO) and bilirubin on CH biosynthesis, CH efflux and oxysterol formation in cultured astroglia. Oxysterols 175-184 heme oxygenase 1 Homo sapiens 51-55 18983509-0 2009 15-deoxy-Delta12,14 prostaglandin J2-induced heme oxygenase-1 in megakaryocytes regulates thrombopoiesis. 9-deoxy-delta-9-prostaglandin D2 20-36 heme oxygenase 1 Homo sapiens 45-61 18983509-10 2009 15d-PGJ(2)-induced HO-1 was independent of PPARgamma, but could be replicated using other electrophilic prostaglandins, suggesting that the electrophilic properties of 15d-PGJ(2) were important for HO-1 induction. 15d-pgj 0-7 heme oxygenase 1 Homo sapiens 19-23 18983509-10 2009 15d-PGJ(2)-induced HO-1 was independent of PPARgamma, but could be replicated using other electrophilic prostaglandins, suggesting that the electrophilic properties of 15d-PGJ(2) were important for HO-1 induction. 15d-pgj 0-7 heme oxygenase 1 Homo sapiens 198-202 18983509-10 2009 15d-PGJ(2)-induced HO-1 was independent of PPARgamma, but could be replicated using other electrophilic prostaglandins, suggesting that the electrophilic properties of 15d-PGJ(2) were important for HO-1 induction. 15d-pgj 168-175 heme oxygenase 1 Homo sapiens 19-23 18983509-11 2009 Interestingly, inhibiting HO-1 activity enhanced ROS generation and augmented 15d-PGJ(2)-induced platelet production, which could be attenuated by antioxidants. Reactive Oxygen Species 49-52 heme oxygenase 1 Homo sapiens 26-30 18983509-11 2009 Interestingly, inhibiting HO-1 activity enhanced ROS generation and augmented 15d-PGJ(2)-induced platelet production, which could be attenuated by antioxidants. 15-deoxyprostaglandin J2 78-88 heme oxygenase 1 Homo sapiens 26-30 18983509-12 2009 These new data reveal that HO-1 negatively regulates thrombopoiesis by inhibiting ROS. Reactive Oxygen Species 82-85 heme oxygenase 1 Homo sapiens 27-31 19468257-0 2009 Pretreatment with glutamine attenuates anoxia/reoxygenation injury of human proximal renal tubular epithelial cells via induction of heme oxygenase-1. Glutamine 18-27 heme oxygenase 1 Homo sapiens 133-149 19468257-1 2009 BACKGROUND/AIMS: Glutamine showed potential cytoprotective activity probably due to induction of heme oxygenase-1 (HO-1). Glutamine 17-26 heme oxygenase 1 Homo sapiens 97-113 19468257-1 2009 BACKGROUND/AIMS: Glutamine showed potential cytoprotective activity probably due to induction of heme oxygenase-1 (HO-1). Glutamine 17-26 heme oxygenase 1 Homo sapiens 115-119 19307691-10 2009 We close this review by focusing on the protective properties of the organic nitrate pentaerithrityl tetranitrate, which upregulates enzymes that have strong antioxidative activity, such as heme oxygenase-1 and ferritin, thereby preventing the development of tolerance and endothelial dysfunction. organic 69-76 heme oxygenase 1 Homo sapiens 190-219 19468257-5 2009 RESULTS: A/R significantly induced apoptosis of HK-2 cells, which was inhibited by glutamine, during which HO-1 was upregulated significantly, but nullified by p38MAPK inhibitor. Glutamine 83-92 heme oxygenase 1 Homo sapiens 107-111 19468257-8 2009 CONCLUSION: Glutamine prevents A/R injury of HK-2 cells through induction of HO-1 via a p38MAPK-dependent pathway, which plays a key role in the cytoprotective effect of glutamine. Glutamine 12-21 heme oxygenase 1 Homo sapiens 77-81 19307691-10 2009 We close this review by focusing on the protective properties of the organic nitrate pentaerithrityl tetranitrate, which upregulates enzymes that have strong antioxidative activity, such as heme oxygenase-1 and ferritin, thereby preventing the development of tolerance and endothelial dysfunction. nitrate pentaerithrityl tetranitrate 77-113 heme oxygenase 1 Homo sapiens 190-219 19468257-8 2009 CONCLUSION: Glutamine prevents A/R injury of HK-2 cells through induction of HO-1 via a p38MAPK-dependent pathway, which plays a key role in the cytoprotective effect of glutamine. Glutamine 170-179 heme oxygenase 1 Homo sapiens 77-81 18957281-9 2008 Thus, PGD(2) induces HO-1 mRNA expression through DP2 receptor, linking the PGD(2)-DP2 signaling with heme homeostasis. Prostaglandin D2 6-12 heme oxygenase 1 Homo sapiens 21-25 18923065-5 2009 However, the YC-1-mediated induction of HO-1 was inhibited by the phosphatidylinositol-3-kinase (PI3K) inhibitors wortmannin and 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002). Wortmannin 114-124 heme oxygenase 1 Homo sapiens 40-44 18923065-5 2009 However, the YC-1-mediated induction of HO-1 was inhibited by the phosphatidylinositol-3-kinase (PI3K) inhibitors wortmannin and 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002). LY-294,002 hydrochloride 129-192 heme oxygenase 1 Homo sapiens 40-44 18923065-5 2009 However, the YC-1-mediated induction of HO-1 was inhibited by the phosphatidylinositol-3-kinase (PI3K) inhibitors wortmannin and 2-(4-morpholinyl)-8-phenyl-1(4H)-benzopyran-4-one hydrochloride (LY294002). 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 194-202 heme oxygenase 1 Homo sapiens 40-44 18923065-8 2009 The ability of YC-1 to sensitize sGC to gaseous monoxides and simultaneously stimulate their production through the induction of HO-1 and iNOS provides a potent mechanism by which the cGMP-dependent and -independent biological actions of this agent are amplified. Cyclic GMP 184-188 heme oxygenase 1 Homo sapiens 129-133 18829152-4 2009 When N-acetyl-L-cysteine or glutathione-monoethyl ester, a potent antioxidant, was added to cell culture, hmox-1 upregulation was attenuated, suggesting that oxidative stress caused by sonication is involved in this process. Acetylcysteine 5-24 heme oxygenase 1 Homo sapiens 106-112 18829152-4 2009 When N-acetyl-L-cysteine or glutathione-monoethyl ester, a potent antioxidant, was added to cell culture, hmox-1 upregulation was attenuated, suggesting that oxidative stress caused by sonication is involved in this process. S-ethyl glutathione 28-55 heme oxygenase 1 Homo sapiens 106-112 18957281-0 2008 Prostaglandin D2 induces heme oxygenase-1 mRNA expression through the DP2 receptor. Prostaglandin D2 0-16 heme oxygenase 1 Homo sapiens 25-41 18957281-2 2008 We have shown that PGD(2) induces the expression of heme oxygenase-1 (HO-1) in the retinal pigment epithelium (RPE) that is essential for survival of photoreceptors. Prostaglandin D2 19-25 heme oxygenase 1 Homo sapiens 52-68 18957281-2 2008 We have shown that PGD(2) induces the expression of heme oxygenase-1 (HO-1) in the retinal pigment epithelium (RPE) that is essential for survival of photoreceptors. Prostaglandin D2 19-25 heme oxygenase 1 Homo sapiens 70-74 18957281-9 2008 Thus, PGD(2) induces HO-1 mRNA expression through DP2 receptor, linking the PGD(2)-DP2 signaling with heme homeostasis. Prostaglandin D2 76-82 heme oxygenase 1 Homo sapiens 21-25 18957281-3 2008 HO-1 is a key enzyme in physiological heme degradation. Heme 38-42 heme oxygenase 1 Homo sapiens 0-4 18845176-7 2008 Moreover, puerarin induced Nrf2 nuclear translocation, which is upstream of puerarin-induced HO-1 expression, and PI3K activation, a pathway that is involved in induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. puerarin 10-18 heme oxygenase 1 Homo sapiens 93-97 18957281-4 2008 Here, we explored the mechanism for the PGD(2)-mediated induction of HO-1 expression using ARPE-19 human RPE cells. Prostaglandin D2 40-46 heme oxygenase 1 Homo sapiens 69-73 18957281-6 2008 Treatment with a DP2 agonist, 15(R)-15-methyl-PGD(2) or DK-PGD(2), increased HO-1 mRNA expression, and pretreatment with a DP2 antagonist, CAY10471, decreased the magnitude of the PGD(2)-mediated HO-1 induction. 15(r)-15-methyl-pgd 30-49 heme oxygenase 1 Homo sapiens 77-81 18957281-6 2008 Treatment with a DP2 agonist, 15(R)-15-methyl-PGD(2) or DK-PGD(2), increased HO-1 mRNA expression, and pretreatment with a DP2 antagonist, CAY10471, decreased the magnitude of the PGD(2)-mediated HO-1 induction. 15(r)-15-methyl-pgd 30-49 heme oxygenase 1 Homo sapiens 196-200 18957281-6 2008 Treatment with a DP2 agonist, 15(R)-15-methyl-PGD(2) or DK-PGD(2), increased HO-1 mRNA expression, and pretreatment with a DP2 antagonist, CAY10471, decreased the magnitude of the PGD(2)-mediated HO-1 induction. Prostaglandins D 46-49 heme oxygenase 1 Homo sapiens 77-81 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. puerarin 53-61 heme oxygenase 1 Homo sapiens 169-185 18845176-7 2008 Moreover, puerarin induced Nrf2 nuclear translocation, which is upstream of puerarin-induced HO-1 expression, and PI3K activation, a pathway that is involved in induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. puerarin 10-18 heme oxygenase 1 Homo sapiens 197-201 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. puerarin 53-61 heme oxygenase 1 Homo sapiens 187-191 18845176-7 2008 Moreover, puerarin induced Nrf2 nuclear translocation, which is upstream of puerarin-induced HO-1 expression, and PI3K activation, a pathway that is involved in induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. puerarin 76-84 heme oxygenase 1 Homo sapiens 93-97 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. tert-Butylhydroperoxide 71-95 heme oxygenase 1 Homo sapiens 169-185 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. tert-Butylhydroperoxide 71-95 heme oxygenase 1 Homo sapiens 187-191 18845176-8 2008 Puerarin-induced up-regulation of HO-1 and cytoprotection against t-BHP were abolished by silencing Nrf2 expression with specific siRNA. puerarin 0-8 heme oxygenase 1 Homo sapiens 34-38 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. tert-Butylhydroperoxide 97-102 heme oxygenase 1 Homo sapiens 169-185 18845176-9 2008 Also, puerarin-mediated increases in PI3K activation and HO-1 induction were reversed by co-treatment with ICI 182,780 and pertussis toxin. puerarin 6-14 heme oxygenase 1 Homo sapiens 57-61 18845176-4 2008 In this study, we demonstrate that the phytoestrogen puerarin inhibits tert-butyl hydroperoxide (t-BHP)-induced oxidative injury via an ER-dependent Gbeta1/PI3K/Akt and heme oxygenase-1 (HO-1) pathway. tert-Butylhydroperoxide 97-102 heme oxygenase 1 Homo sapiens 187-191 18845176-10 2008 Taken together, these results suggest that puerarin augments cellular antioxidant defense capacity through ER-dependent HO-1 induction via the Gbeta1/PI3K/Akt-Nrf2 signaling pathway, thereby protecting cells from oxidative stress. puerarin 43-51 heme oxygenase 1 Homo sapiens 120-124 18845176-6 2008 Also, puerarin up-regulated HO-1 expression and this expression conferred cytoprotection against oxidative injury induced by t-BHP. tert-Butylhydroperoxide 125-130 heme oxygenase 1 Homo sapiens 28-32 18852027-0 2008 Nordihydroguaiaretic acid activates the antioxidant pathway Nrf2/HO-1 and protects cerebellar granule neurons against oxidative stress. Masoprocol 0-25 heme oxygenase 1 Homo sapiens 65-69 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Masoprocol 14-39 heme oxygenase 1 Homo sapiens 129-150 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Masoprocol 14-39 heme oxygenase 1 Homo sapiens 152-156 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Masoprocol 41-45 heme oxygenase 1 Homo sapiens 129-150 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Masoprocol 41-45 heme oxygenase 1 Homo sapiens 152-156 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Water 214-219 heme oxygenase 1 Homo sapiens 129-150 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. Water 214-219 heme oxygenase 1 Homo sapiens 152-156 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. 3-nitropropionic acid 226-247 heme oxygenase 1 Homo sapiens 129-150 18852027-2 2008 We found that nordihydroguaiaretic acid (NDGA), a powerful antioxidant from Larrea tridentate, activates the antioxidant pathway Nrf2/heme oxygenase-1 (HO-1) in cerebellar granule neurons and protects them against H(2)O(2) or 3-nitropropionic acid-induced neurotoxicity. 3-nitropropionic acid 226-247 heme oxygenase 1 Homo sapiens 152-156 18852027-3 2008 The role of HO-1 in this protective effect was made evident by using an HO inhibitor, tin-mesoporphyrin. tin mesoporphyrin 86-103 heme oxygenase 1 Homo sapiens 12-16 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. Prednisolone 128-140 heme oxygenase 1 Homo sapiens 299-303 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. Dexamethasone 146-159 heme oxygenase 1 Homo sapiens 38-42 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. 11,17-dihydroxy-6-methyl-17-(1-propynyl)androsta-1,4,6-triene-3-one 119-126 heme oxygenase 1 Homo sapiens 38-42 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. Acetylcysteine 175-194 heme oxygenase 1 Homo sapiens 38-42 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. 11,17-dihydroxy-6-methyl-17-(1-propynyl)androsta-1,4,6-triene-3-one 119-126 heme oxygenase 1 Homo sapiens 299-303 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. Prednisolone 128-140 heme oxygenase 1 Homo sapiens 38-42 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. Acetylcysteine 175-194 heme oxygenase 1 Homo sapiens 299-303 19548360-5 2008 The increases in reactive species and HO-1 protein are inhibited by agonists of glucocorticoid receptors (GR), such as RU28362, prednisolone, and dexamethasone, as well as by N-acetyl-L-cysteine and SB203580 (a p38 inhibitor), suggesting a role of GR in NF-induced increases in reactive species and HO-1. SB 203580 199-207 heme oxygenase 1 Homo sapiens 38-42 19548360-6 2008 Assay-guided fractionation of NF led to three active compounds, phenethyl ferulate, bergaptol, and isoimperatorin, that were found to increase oxidative stress and HO-1 protein levels in HFHs. phenethyl ferulate 64-82 heme oxygenase 1 Homo sapiens 164-168 19548360-6 2008 Assay-guided fractionation of NF led to three active compounds, phenethyl ferulate, bergaptol, and isoimperatorin, that were found to increase oxidative stress and HO-1 protein levels in HFHs. bergaptol 84-93 heme oxygenase 1 Homo sapiens 164-168 19548360-6 2008 Assay-guided fractionation of NF led to three active compounds, phenethyl ferulate, bergaptol, and isoimperatorin, that were found to increase oxidative stress and HO-1 protein levels in HFHs. isoimperatorin 99-113 heme oxygenase 1 Homo sapiens 164-168 18341478-8 2008 SnP (tin protoporphyrin), an HO-1 inhibitor, counteracted the effects of Adv-HO-1. tin protoporphyrin IX 5-23 heme oxygenase 1 Homo sapiens 29-33 18341478-8 2008 SnP (tin protoporphyrin), an HO-1 inhibitor, counteracted the effects of Adv-HO-1. tin protoporphyrin IX 5-23 heme oxygenase 1 Homo sapiens 77-81 19120903-2 2008 Two end products of heme degradation, carbon monoxide (CO) and bilirubin, are involved in the protective role of HO-1 against oxidative injury. Carbon Monoxide 38-53 heme oxygenase 1 Homo sapiens 113-117 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Cadmium 152-154 heme oxygenase 1 Homo sapiens 182-198 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Cadmium 152-154 heme oxygenase 1 Homo sapiens 200-204 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Zinc 243-245 heme oxygenase 1 Homo sapiens 200-204 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Manganese 254-263 heme oxygenase 1 Homo sapiens 200-204 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Cadmium 322-324 heme oxygenase 1 Homo sapiens 200-204 18948096-4 2008 The present study had a focus on the human retinal pigment epithelial (RPE) cell line ARPE-19 in an attempt to demonstrate a reduction in intracellular Cd(2+) effect associated with heme oxygenase-1 (HO-1) expression by co-exposure with zinc (Zn(2+)) or manganese (Mn(2+)), which is known to be a more potent inhibitor of Cd(2+) uptake than Zn(2+). Zinc 341-343 heme oxygenase 1 Homo sapiens 200-204 18948096-7 2008 Mn(2+) ions as low as 2.5 microM were found to cause an increase in HO-1 mRNA expression levels in ARPE-19 cells, demonstrating for the first time that Mn(2+) is an inducer of HO-1. Manganese(2+) 0-6 heme oxygenase 1 Homo sapiens 68-72 18948096-7 2008 Mn(2+) ions as low as 2.5 microM were found to cause an increase in HO-1 mRNA expression levels in ARPE-19 cells, demonstrating for the first time that Mn(2+) is an inducer of HO-1. Manganese(2+) 0-6 heme oxygenase 1 Homo sapiens 176-180 18948096-7 2008 Mn(2+) ions as low as 2.5 microM were found to cause an increase in HO-1 mRNA expression levels in ARPE-19 cells, demonstrating for the first time that Mn(2+) is an inducer of HO-1. Manganese(2+) 152-158 heme oxygenase 1 Homo sapiens 68-72 18948096-7 2008 Mn(2+) ions as low as 2.5 microM were found to cause an increase in HO-1 mRNA expression levels in ARPE-19 cells, demonstrating for the first time that Mn(2+) is an inducer of HO-1. Manganese(2+) 152-158 heme oxygenase 1 Homo sapiens 176-180 18948096-8 2008 Mn(2+) ions at 1 microM induced HO-1 mRNA expression in the HEK293 human embryonic kidney cells. Manganese(2+) 0-6 heme oxygenase 1 Homo sapiens 32-36 18948096-11 2008 Further, induction of HO-1 by Mn(2+) could provide RPE with some resistance to enhanced oxidative stress arising from Cd(2+) accumulation in RPE as HO-1 is one of the frontline cellular antioxidant defense mechanisms. Manganese(2+) 30-36 heme oxygenase 1 Homo sapiens 22-26 18948096-11 2008 Further, induction of HO-1 by Mn(2+) could provide RPE with some resistance to enhanced oxidative stress arising from Cd(2+) accumulation in RPE as HO-1 is one of the frontline cellular antioxidant defense mechanisms. Manganese(2+) 30-36 heme oxygenase 1 Homo sapiens 148-152 18301921-2 2008 The enzyme heme oxygenase-1 (HO-1) participates in cytoprotection against oxygen radical injury. Oxygen 16-22 heme oxygenase 1 Homo sapiens 29-33 19120903-2 2008 Two end products of heme degradation, carbon monoxide (CO) and bilirubin, are involved in the protective role of HO-1 against oxidative injury. Carbon Monoxide 55-57 heme oxygenase 1 Homo sapiens 113-117 19120903-2 2008 Two end products of heme degradation, carbon monoxide (CO) and bilirubin, are involved in the protective role of HO-1 against oxidative injury. Bilirubin 63-72 heme oxygenase 1 Homo sapiens 113-117 18799798-0 2008 Overexpression of heme oxygenase-1 protects dopaminergic neurons against 1-methyl-4-phenylpyridinium-induced neurotoxicity. 1-Methyl-4-phenylpyridinium 73-100 heme oxygenase 1 Homo sapiens 18-34 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Heme 117-121 heme oxygenase 1 Homo sapiens 0-16 18799798-6 2008 Apomorphine-induced rotation after MPP(+) treatment was also inhibited by Ad-HO-1. Apomorphine 0-11 heme oxygenase 1 Homo sapiens 77-81 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Heme 117-121 heme oxygenase 1 Homo sapiens 18-22 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 0-16 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Carbon Monoxide 125-140 heme oxygenase 1 Homo sapiens 18-22 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 0-16 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Carbon Monoxide 142-144 heme oxygenase 1 Homo sapiens 18-22 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Iron 147-151 heme oxygenase 1 Homo sapiens 0-16 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Iron 147-151 heme oxygenase 1 Homo sapiens 18-22 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Bilirubin 157-166 heme oxygenase 1 Homo sapiens 0-16 18799798-1 2008 Heme oxygenase-1 (HO-1) is up-regulated in response to oxidative stress and catalyzes the degradation of pro-oxidant heme to carbon monoxide (CO), iron, and bilirubin. Bilirubin 157-166 heme oxygenase 1 Homo sapiens 18-22 18799798-3 2008 We here locally injected adenovirus containing human HO-1 gene (Ad-HO-1) into rat substantia nigra concomitantly with 1-methyl-4-phenylpyridinium (MPP(+)). 1-Methyl-4-phenylpyridinium 118-145 heme oxygenase 1 Homo sapiens 53-57 18799798-3 2008 We here locally injected adenovirus containing human HO-1 gene (Ad-HO-1) into rat substantia nigra concomitantly with 1-methyl-4-phenylpyridinium (MPP(+)). 1-Methyl-4-phenylpyridinium 118-145 heme oxygenase 1 Homo sapiens 67-71 18799798-3 2008 We here locally injected adenovirus containing human HO-1 gene (Ad-HO-1) into rat substantia nigra concomitantly with 1-methyl-4-phenylpyridinium (MPP(+)). mangion-purified polysaccharide (Candida albicans) 147-150 heme oxygenase 1 Homo sapiens 53-57 18809379-0 2008 NS-398, a selective COX-2 inhibitor, inhibits proliferation of IL-1beta-stimulated vascular smooth muscle cells by induction of HO-1. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 0-6 heme oxygenase 1 Homo sapiens 128-132 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. Dinoprostone 73-79 heme oxygenase 1 Homo sapiens 142-146 18809379-1 2008 We investigated whether NS-398, a selective inhibitor of COX-2, induces HO-1 in IL-1beta-stimulated vascular smooth muscle cells (VSMC). N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 24-30 heme oxygenase 1 Homo sapiens 72-76 18809379-5 2008 Hemin, a HO-1 inducer, also reduced the production of PGE(2) and proliferation of IL-1beta-stimulated VSMC. Prostaglandins E 54-57 heme oxygenase 1 Homo sapiens 9-13 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. tin protoporphyrin IX 13-19 heme oxygenase 1 Homo sapiens 23-27 18809379-8 2008 In conclusion, NS-398 inhibits proliferation of IL-1beta-stimulated VSMC by HO-1-derived CO. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 15-21 heme oxygenase 1 Homo sapiens 76-80 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. tin protoporphyrin IX 13-19 heme oxygenase 1 Homo sapiens 142-146 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 63-69 heme oxygenase 1 Homo sapiens 23-27 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 63-69 heme oxygenase 1 Homo sapiens 142-146 18809379-4 2008 Furthermore, SnPPIX, a HO-1 inhibitor, reversed the effects of NS-398 on PGE(2) production, suggesting that COX-2 activity can be affected by HO-1. Dinoprostone 73-79 heme oxygenase 1 Homo sapiens 23-27 18696091-12 2008 Both cell types biosynthesize HO-1 and ferritin in response to heme. Heme 63-67 heme oxygenase 1 Homo sapiens 30-34 18528644-10 2008 Heme oxygenase 1 expression increased and DMT1 expression decreased with higher heme Fe concentrations in the media. Heme 80-84 heme oxygenase 1 Homo sapiens 0-16 18528644-10 2008 Heme oxygenase 1 expression increased and DMT1 expression decreased with higher heme Fe concentrations in the media. Iron 85-87 heme oxygenase 1 Homo sapiens 0-16 18928848-3 2008 In addition, treatment with N-acetylcysteine, indomethacin, and heme oxygenase-1 inhibitors blocked reactive oxygen species production, antioxidant response element (ARE) gene expression, and Nrf2 accumulation that occurred in response to mechanical stress. Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 64-80 18778734-10 2008 While induction of heme oxygenase-1 was implicated in the cytoprotection by 15d-PGJ(2) under some experimental conditions, additional experiments indicated that this enzyme was not involved in the cytoprotection observed in this system. 15d-pgj 76-83 heme oxygenase 1 Homo sapiens 19-35 18985801-6 2008 However, in recent years a possible novel aspect in the mode of action of these compounds has been suggested; that is, the ultimate stimulation of the heme oxygenase-1 (HO-1) pathway is likely to account for the established and powerful antioxidant/anti-inflammatory properties of these polyphenols. Polyphenols 287-298 heme oxygenase 1 Homo sapiens 151-167 18985801-6 2008 However, in recent years a possible novel aspect in the mode of action of these compounds has been suggested; that is, the ultimate stimulation of the heme oxygenase-1 (HO-1) pathway is likely to account for the established and powerful antioxidant/anti-inflammatory properties of these polyphenols. Polyphenols 287-298 heme oxygenase 1 Homo sapiens 169-173 18798608-0 2008 X-ray crystal structure of human heme oxygenase-1 in complex with 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone: a common binding mode for imidazole-based heme oxygenase-1 inhibitors. 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone 66-113 heme oxygenase 1 Homo sapiens 33-49 18798608-0 2008 X-ray crystal structure of human heme oxygenase-1 in complex with 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone: a common binding mode for imidazole-based heme oxygenase-1 inhibitors. 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone 66-113 heme oxygenase 1 Homo sapiens 157-173 18798608-0 2008 X-ray crystal structure of human heme oxygenase-1 in complex with 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone: a common binding mode for imidazole-based heme oxygenase-1 inhibitors. imidazole 141-150 heme oxygenase 1 Homo sapiens 33-49 18798608-0 2008 X-ray crystal structure of human heme oxygenase-1 in complex with 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethanone: a common binding mode for imidazole-based heme oxygenase-1 inhibitors. imidazole 141-150 heme oxygenase 1 Homo sapiens 157-173 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. guanine-thymidine 15-32 heme oxygenase 1 Homo sapiens 53-58 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. guanine-thymidine 15-32 heme oxygenase 1 Homo sapiens 131-147 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. guanine-thymidine 15-32 heme oxygenase 1 Homo sapiens 149-153 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. Heme 109-113 heme oxygenase 1 Homo sapiens 53-58 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. Heme 109-113 heme oxygenase 1 Homo sapiens 131-147 18975324-1 2008 OBJECTIVE: The guanine-thymidine (GT)n repeat in the HMOX1 promoter determines the level of induction of the heme-degrading enzyme heme oxygenase 1 (HO-1), which protects against inflammatory and oxidative stress. Heme 109-113 heme oxygenase 1 Homo sapiens 149-153 18824663-3 2008 Although the HO-1 gene promoter contains consensus binding sites for proinflammatory/oxidative transcription factors like nuclear factor-kappaB, activating protein (AP)-1, and AP-2, the effects of HO inducers on these transcription factors in cardiac lesions of deoxycorticosterone acetate hypertension are not fully understood. Desoxycorticosterone Acetate 262-289 heme oxygenase 1 Homo sapiens 13-17 18824663-5 2008 The cardioprotection by hemin was accompanied by increased HO-1, HO activity, cGMP, superoxide dismutase, catalase, the total antioxidant capacity alongside the reduction of 8-isoprostane, AP-1, AP-2, nuclear factor-kappaB, and c-Jun-NH(2)-terminal kinase, whereas chromium mesoporphyrin abolished the hemin effects. Hemin 24-29 heme oxygenase 1 Homo sapiens 59-63 18799519-0 2008 A prolyl-hydroxylase inhibitor, ethyl-3,4-dihydroxybenzoate, induces haem oxygenase-1 expression in human cells through a mechanism independent of hypoxia-inducible factor-1alpha. ethyl protocatechuate 32-59 heme oxygenase 1 Homo sapiens 69-85 18799519-4 2008 Under hypoxia (1% O(2)), the expression of HO-1 mRNA was decreased in HeLa cells, increased in D407 cells, and unchanged in ARPE-19 cells, while HIF-1alpha protein was accumulated in these cell lines. o(2) 18-22 heme oxygenase 1 Homo sapiens 43-47 18799519-7 2008 Treatment with EDHB (250-500 microM) increased HIF-1alpha protein levels in HeLa and D407 cells, but not in ARPE-19 cells, whereas EDHB at lower concentrations (50-100 microM) consistently induced HO-1 mRNA expression (about 20-fold) in these three cell lines. ethyl protocatechuate 15-19 heme oxygenase 1 Homo sapiens 197-201 18799519-7 2008 Treatment with EDHB (250-500 microM) increased HIF-1alpha protein levels in HeLa and D407 cells, but not in ARPE-19 cells, whereas EDHB at lower concentrations (50-100 microM) consistently induced HO-1 mRNA expression (about 20-fold) in these three cell lines. ethyl protocatechuate 131-135 heme oxygenase 1 Homo sapiens 197-201 18799519-8 2008 Moreover, EDHB increased the HO-1 gene promoter activity via the enhancer that lacks a HIF-1-binding site. ethyl protocatechuate 10-14 heme oxygenase 1 Homo sapiens 29-33 18799519-9 2008 In conclusion, the signals evoked by hypoxia and after EDHB treatment differentially regulate HO-1 mRNA expression through HIF-1alpha-independent mechanisms. ethyl protocatechuate 55-59 heme oxygenase 1 Homo sapiens 94-98 18524839-10 2008 A stronger estimated effect of O(3) on FEV(1) was found in subjects carrying both the GSTP1 105Val variant and the HMOX1 long (GT)n repeat (-1.94%, 95% CI: -2.89% to -0.98%). Ozone 31-35 heme oxygenase 1 Homo sapiens 115-120 18787101-1 2008 We investigated the role of heme oxygenase-1 (HO-1), a powerful anti-inflammatory and anti-oxidant enzyme, in modulating cigarette smoke (CS)-induced mucus secretion. Cesium 138-140 heme oxygenase 1 Homo sapiens 28-44 18787101-1 2008 We investigated the role of heme oxygenase-1 (HO-1), a powerful anti-inflammatory and anti-oxidant enzyme, in modulating cigarette smoke (CS)-induced mucus secretion. Cesium 138-140 heme oxygenase 1 Homo sapiens 46-50 18787101-4 2008 In French participants to the European Community Respiratory Health Survey II (n = 210, 30 to 53 years of age, 50% males) exposed to CS, a significant increase in the percentage of participants with chronic sputum was observed in those harboring at least one allele with a long (GT)(n) in the HO-1 promoter gene (>33 repeats), which is associated with a low level of HO-1 protein expression, compared with those with a short number of (GT)n repeats (21.7% versus 8.6%, P = 0.047). Cesium 133-135 heme oxygenase 1 Homo sapiens 293-297 18787101-4 2008 In French participants to the European Community Respiratory Health Survey II (n = 210, 30 to 53 years of age, 50% males) exposed to CS, a significant increase in the percentage of participants with chronic sputum was observed in those harboring at least one allele with a long (GT)(n) in the HO-1 promoter gene (>33 repeats), which is associated with a low level of HO-1 protein expression, compared with those with a short number of (GT)n repeats (21.7% versus 8.6%, P = 0.047). Cesium 133-135 heme oxygenase 1 Homo sapiens 370-374 18787101-6 2008 We conclude that HO-1 has a significant protective effect against airway mucus hypersecretion in animals and humans exposed to CS. Cesium 127-129 heme oxygenase 1 Homo sapiens 17-21 18696091-14 2008 The persistence of HO-1 protein implies continuous exposure of CNS to free heme or an excessively sensitive transcriptional response of the HO-1 gene. Heme 75-79 heme oxygenase 1 Homo sapiens 19-23 18701634-5 2008 HO-1 was induced in cultured mTALH cells by treatment with cobalt protoporphyrin (CoPP, 10 microM) or hemin (50 microM) or by transfection with a plasmid containing the human HO-1 isoform. cobaltiprotoporphyrin 59-80 heme oxygenase 1 Homo sapiens 0-4 18689604-8 2008 Treatment with the antioxidants N-acetyl-l-cysteine or GSH reduced the expression of HO-1 induced by CSE. Glutathione 55-58 heme oxygenase 1 Homo sapiens 85-89 18689604-0 2008 Cigarette smoke-induced expression of heme oxygenase-1 in human lung fibroblasts is regulated by intracellular glutathione. Glutathione 111-122 heme oxygenase 1 Homo sapiens 38-54 18701634-5 2008 HO-1 was induced in cultured mTALH cells by treatment with cobalt protoporphyrin (CoPP, 10 microM) or hemin (50 microM) or by transfection with a plasmid containing the human HO-1 isoform. cobaltiprotoporphyrin 82-86 heme oxygenase 1 Homo sapiens 0-4 18689604-7 2008 This induction of HO-1 paralleled a decrease in intracellular GSH, and a sustained reduction in GSH resulted in a dramatic increase in HO-1. Glutathione 62-65 heme oxygenase 1 Homo sapiens 18-22 18689604-13 2008 Reduction of c-Fos and c-Jun nuclear translocation by SP-600125 attenuated the CSE-induced expression of HO-1. pyrazolanthrone 54-63 heme oxygenase 1 Homo sapiens 105-109 18701634-5 2008 HO-1 was induced in cultured mTALH cells by treatment with cobalt protoporphyrin (CoPP, 10 microM) or hemin (50 microM) or by transfection with a plasmid containing the human HO-1 isoform. Hemin 102-107 heme oxygenase 1 Homo sapiens 0-4 18689604-7 2008 This induction of HO-1 paralleled a decrease in intracellular GSH, and a sustained reduction in GSH resulted in a dramatic increase in HO-1. Glutathione 96-99 heme oxygenase 1 Homo sapiens 135-139 18689604-8 2008 Treatment with the antioxidants N-acetyl-l-cysteine or GSH reduced the expression of HO-1 induced by CSE. Acetylcysteine 32-51 heme oxygenase 1 Homo sapiens 85-89 18701634-7 2008 Induction of HO-1 via CoPP, hemin, or overexpression of the human HO-1 isoform significantly reduced ANG II-induced DHE fluorescence to 64+/-5, 64+/-8, and 41+/-4 RFU/microm2, respectively. dihydroethidium 116-119 heme oxygenase 1 Homo sapiens 13-17 18701634-7 2008 Induction of HO-1 via CoPP, hemin, or overexpression of the human HO-1 isoform significantly reduced ANG II-induced DHE fluorescence to 64+/-5, 64+/-8, and 41+/-4 RFU/microm2, respectively. dihydroethidium 116-119 heme oxygenase 1 Homo sapiens 66-70 18544348-6 2008 HO-1 ubiquitination in HEK293 cells was enhanced by zinc chloride, but suppressed with a zinc chelator (N,N,N",N"-tetrakis(2-pyridylmethyl)ethyl-enediamine), suggesting the involvement of a RING-E3 ligase in this process. zinc chloride 52-65 heme oxygenase 1 Homo sapiens 0-4 18576916-1 2008 Heme oxygenase-1, an enzyme degrading heme to carbon monoxide, iron, and biliverdin, has been recognized as playing a crucial role in cellular defense against stressful conditions, not only related to heme release. Heme 38-42 heme oxygenase 1 Homo sapiens 0-16 18576916-1 2008 Heme oxygenase-1, an enzyme degrading heme to carbon monoxide, iron, and biliverdin, has been recognized as playing a crucial role in cellular defense against stressful conditions, not only related to heme release. Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 0-16 18576916-1 2008 Heme oxygenase-1, an enzyme degrading heme to carbon monoxide, iron, and biliverdin, has been recognized as playing a crucial role in cellular defense against stressful conditions, not only related to heme release. Iron 63-67 heme oxygenase 1 Homo sapiens 0-16 18576916-1 2008 Heme oxygenase-1, an enzyme degrading heme to carbon monoxide, iron, and biliverdin, has been recognized as playing a crucial role in cellular defense against stressful conditions, not only related to heme release. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 0-16 18576916-1 2008 Heme oxygenase-1, an enzyme degrading heme to carbon monoxide, iron, and biliverdin, has been recognized as playing a crucial role in cellular defense against stressful conditions, not only related to heme release. Heme 201-205 heme oxygenase 1 Homo sapiens 0-16 18769149-13 2008 Conversely, transfection of Beas-2b with ho-1 siRNA augmented CSE-induced DISC formation and increased intracellular reactive oxygen species formation. Reactive Oxygen Species 117-140 heme oxygenase 1 Homo sapiens 41-45 18769232-1 2008 PURPOSE OF REVIEW: Heme oxygenase-1 apart from converting heme to carbon monoxide, iron and biliverdin has been shown to exert anti-inflammatory, antiapoptotic and antioxidant actions. Heme 58-62 heme oxygenase 1 Homo sapiens 19-35 18769232-1 2008 PURPOSE OF REVIEW: Heme oxygenase-1 apart from converting heme to carbon monoxide, iron and biliverdin has been shown to exert anti-inflammatory, antiapoptotic and antioxidant actions. Carbon Monoxide 66-81 heme oxygenase 1 Homo sapiens 19-35 18769232-1 2008 PURPOSE OF REVIEW: Heme oxygenase-1 apart from converting heme to carbon monoxide, iron and biliverdin has been shown to exert anti-inflammatory, antiapoptotic and antioxidant actions. Iron 83-87 heme oxygenase 1 Homo sapiens 19-35 18769232-1 2008 PURPOSE OF REVIEW: Heme oxygenase-1 apart from converting heme to carbon monoxide, iron and biliverdin has been shown to exert anti-inflammatory, antiapoptotic and antioxidant actions. Biliverdine 92-102 heme oxygenase 1 Homo sapiens 19-35 18769232-3 2008 RECENT FINDINGS: Heme oxygenase-1 has been shown to be protective against atherosclerosis via decreasing ROS generation and proinflammatory cytokine production resulting in diminished lipid uptake and foam cell formation. ros 105-108 heme oxygenase 1 Homo sapiens 17-33 18802114-0 2008 Lipoteichoic acid induces HO-1 expression via the TLR2/MyD88/c-Src/NADPH oxidase pathway and Nrf2 in human tracheal smooth muscle cells. lipoteichoic acid 0-17 heme oxygenase 1 Homo sapiens 26-30 18802114-1 2008 Heme oxygenase (HO)-1 is a stress-inducible rate-limiting enzyme in heme degradation that confers cytoprotection against oxidative injury and provides a vital function in maintaining tissue homeostasis. Heme 68-72 heme oxygenase 1 Homo sapiens 0-21 18802114-7 2008 These results demonstrated that LTA-induced ROS generation was mediated through the TLR2/MyD88/TRAF6/c-Src/NADPH oxidase pathway, in turn initiates the activation of Nrf2, and ultimately induces HO-1 expression in HTSMCs. Reactive Oxygen Species 44-47 heme oxygenase 1 Homo sapiens 195-199 18689445-6 2008 The application of the HO-1-specific inhibitor zinc protoporphyrin IX (ZnPPIX) could inhibit the above IAA and hematin responses. zinc protoporphyrin 47-69 heme oxygenase 1 Homo sapiens 23-27 18689445-6 2008 The application of the HO-1-specific inhibitor zinc protoporphyrin IX (ZnPPIX) could inhibit the above IAA and hematin responses. zinc protoporphyrin 71-77 heme oxygenase 1 Homo sapiens 23-27 18586083-0 2008 Role of oxidative stress in the induction of metallothionein-2A and heme oxygenase-1 gene expression by the antineoplastic agent gallium nitrate in human lymphoma cells. gallium nitrate 129-144 heme oxygenase 1 Homo sapiens 68-84 18786476-1 2008 Hemoxygenase (HO)-1 is an inducible isoform of the first and rate-controlling enzyme of the degradation of heme into iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Heme 107-111 heme oxygenase 1 Homo sapiens 0-19 18786476-1 2008 Hemoxygenase (HO)-1 is an inducible isoform of the first and rate-controlling enzyme of the degradation of heme into iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Iron 117-121 heme oxygenase 1 Homo sapiens 0-19 18786476-1 2008 Hemoxygenase (HO)-1 is an inducible isoform of the first and rate-controlling enzyme of the degradation of heme into iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Carbon Monoxide 123-138 heme oxygenase 1 Homo sapiens 0-19 18786476-1 2008 Hemoxygenase (HO)-1 is an inducible isoform of the first and rate-controlling enzyme of the degradation of heme into iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Biliverdine 144-154 heme oxygenase 1 Homo sapiens 0-19 18786476-1 2008 Hemoxygenase (HO)-1 is an inducible isoform of the first and rate-controlling enzyme of the degradation of heme into iron, carbon monoxide, and biliverdin, the latter being subsequently converted into bilirubin. Bilirubin 201-210 heme oxygenase 1 Homo sapiens 0-19 18786476-3 2008 Thus, the physiological induction of HO-1 may be an adaptive and beneficial response to several possibly noxious stimuli, including heme itself, suggesting a potentially autoprotective and autodefensive role in several pathophysiological states including acute coronary syndromes and stroke. Heme 132-136 heme oxygenase 1 Homo sapiens 37-41 18566994-4 2008 The activation of Nrf2 by apo-10"-lycopenoic acid is associated with the induction of phase II detoxifying/antioxidant enzymes including heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1, glutathione S-transferases, and glutamate-cysteine ligases in BEAS-2B cells. apo-10'-lycopenoic acid 26-49 heme oxygenase 1 Homo sapiens 137-187 18586083-6 2008 N-Acetyl-L-cysteine blocked gallium-induced MT2A and HO-1 expression and increased gallium"s cytotoxicity. Acetylcysteine 0-19 heme oxygenase 1 Homo sapiens 53-57 18586083-6 2008 N-Acetyl-L-cysteine blocked gallium-induced MT2A and HO-1 expression and increased gallium"s cytotoxicity. Gallium 28-35 heme oxygenase 1 Homo sapiens 53-57 18586083-2 2008 Using a DNA microarray to examine genes induced by gallium nitrate in CCRF-CEM cells, we found that gallium increased metallothionein-2A (MT2A) and heme oxygenase-1 (HO-1) gene expression and altered the levels of other stress-related genes. gallium nitrate 51-66 heme oxygenase 1 Homo sapiens 148-164 18586083-8 2008 Gallium nitrate increased the phosphorylation of p38 mitogen-activated protein kinase and activated Nrf-2, a regulator of HO-1 gene transcription. gallium nitrate 0-15 heme oxygenase 1 Homo sapiens 122-126 18586083-9 2008 Gallium-induced Nrf-2 activation and HO-1 expression were diminished by a p38 MAP kinase inhibitor. Gallium 0-7 heme oxygenase 1 Homo sapiens 37-41 18586083-2 2008 Using a DNA microarray to examine genes induced by gallium nitrate in CCRF-CEM cells, we found that gallium increased metallothionein-2A (MT2A) and heme oxygenase-1 (HO-1) gene expression and altered the levels of other stress-related genes. gallium nitrate 51-66 heme oxygenase 1 Homo sapiens 166-170 18586083-10 2008 We conclude that gallium nitrate induces cellular oxidative stress as an early event which then triggers the expression of HO-1 and MT2A through different pathways. gallium nitrate 17-32 heme oxygenase 1 Homo sapiens 123-127 18586083-2 2008 Using a DNA microarray to examine genes induced by gallium nitrate in CCRF-CEM cells, we found that gallium increased metallothionein-2A (MT2A) and heme oxygenase-1 (HO-1) gene expression and altered the levels of other stress-related genes. Gallium 51-58 heme oxygenase 1 Homo sapiens 148-164 18586083-2 2008 Using a DNA microarray to examine genes induced by gallium nitrate in CCRF-CEM cells, we found that gallium increased metallothionein-2A (MT2A) and heme oxygenase-1 (HO-1) gene expression and altered the levels of other stress-related genes. Gallium 51-58 heme oxygenase 1 Homo sapiens 166-170 18586083-3 2008 MT2A and HO-1 were increased after 6 and 16 h of incubation with gallium nitrate. gallium nitrate 65-80 heme oxygenase 1 Homo sapiens 9-13 18573251-6 2008 Inhibition of HO-1 activity using the HO-1 inhibitor tin protoporphyrin IX (SnPPIX), resulted in loss of cytoprotection. tin protoporphyrin IX 53-74 heme oxygenase 1 Homo sapiens 14-18 18554677-0 2008 Metallothionein-III protects against 6-hydroxydopamine-induced oxidative stress by increasing expression of heme oxygenase-1 in a PI3K and ERK/Nrf2-dependent manner. Oxidopamine 37-54 heme oxygenase 1 Homo sapiens 108-124 18554677-3 2008 In this study, we demonstrate that MT-III prevents the accumulation of reactive oxygen species (ROS) in dopaminergic SH-SY5Y cells challenged with the Parkinson"s disease-related neurotoxin 6-hydroxydopamine (6-OHDA) by a mechanism that involves phosphatidylinositol 3-kinase (PI3K) and ERK kinase/NF-E2-related factor 2 (Nrf2) dependent induction of the stress response protein heme oxygenase-1 (HO-1). Oxidopamine 190-207 heme oxygenase 1 Homo sapiens 379-395 18554677-3 2008 In this study, we demonstrate that MT-III prevents the accumulation of reactive oxygen species (ROS) in dopaminergic SH-SY5Y cells challenged with the Parkinson"s disease-related neurotoxin 6-hydroxydopamine (6-OHDA) by a mechanism that involves phosphatidylinositol 3-kinase (PI3K) and ERK kinase/NF-E2-related factor 2 (Nrf2) dependent induction of the stress response protein heme oxygenase-1 (HO-1). Oxidopamine 190-207 heme oxygenase 1 Homo sapiens 397-401 18554677-5 2008 Also, MT-III up-regulates HO-1 expression and this expression confers neuroprotection against oxidative injury induced by 6-OHDA. Oxidopamine 122-128 heme oxygenase 1 Homo sapiens 26-30 18554677-7 2008 Taken together, these results suggest that the PI3K and ERK/Nrf2 signaling pathway controls the intracellular levels of ROS by regulating the expression of the antioxidant enzyme HO-1. Reactive Oxygen Species 120-123 heme oxygenase 1 Homo sapiens 179-183 18586007-0 2008 Epigallocatechin activates haem oxygenase-1 expression via protein kinase Cdelta and Nrf2. gallocatechol 0-16 heme oxygenase 1 Homo sapiens 27-43 18586007-4 2008 EGC time and dose dependently increased HO-1 mRNA and protein expression but had minimal effect on expression of other ARE-regulated genes, including NAD(P)H:quinone oxidoreductase 1, glutathione cysteine ligase and ferritin. gallocatechol 0-3 heme oxygenase 1 Homo sapiens 40-44 18586007-5 2008 siRNA knock down of Nrf2 significantly inhibited EGC-induced HO-1 expression. gallocatechol 49-52 heme oxygenase 1 Homo sapiens 61-65 18573251-6 2008 Inhibition of HO-1 activity using the HO-1 inhibitor tin protoporphyrin IX (SnPPIX), resulted in loss of cytoprotection. tin protoporphyrin IX 53-74 heme oxygenase 1 Homo sapiens 38-42 18586007-6 2008 Furthermore, inhibition of PKC by Ro-31-8220 dose dependently decreased EGC-induced HO-1 mRNA expression, whereas MAP kinase and phosphatidylinositol-3-kinase pathway inhibitors had no significant effect. Ro 31-8220 34-44 heme oxygenase 1 Homo sapiens 84-88 18573251-6 2008 Inhibition of HO-1 activity using the HO-1 inhibitor tin protoporphyrin IX (SnPPIX), resulted in loss of cytoprotection. tin protoporphyrin IX 76-82 heme oxygenase 1 Homo sapiens 14-18 18586007-6 2008 Furthermore, inhibition of PKC by Ro-31-8220 dose dependently decreased EGC-induced HO-1 mRNA expression, whereas MAP kinase and phosphatidylinositol-3-kinase pathway inhibitors had no significant effect. gallocatechol 72-75 heme oxygenase 1 Homo sapiens 84-88 18586007-8 2008 PKCdelta inhibition significantly decreased EGC-induced HO-1 mRNA expression, whereas PKCalpha- and beta-specific inhibitors had no significant effect. gallocatechol 44-47 heme oxygenase 1 Homo sapiens 56-60 18586007-9 2008 These results demonstrate for the first time that EGC-induced HO-1 expression occurs via PKCdelta and Nrf2. gallocatechol 50-53 heme oxygenase 1 Homo sapiens 62-66 18573251-6 2008 Inhibition of HO-1 activity using the HO-1 inhibitor tin protoporphyrin IX (SnPPIX), resulted in loss of cytoprotection. tin protoporphyrin IX 76-82 heme oxygenase 1 Homo sapiens 38-42 18573251-7 2008 Carbon monoxide, one of HO-1 catabolic products appeared to play a small role in CAPE protection. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 24-28 18573251-9 2008 These findings suggest an important role of HO-1 induction in CAPE cytoprotection against oxidant stress, which may not relate to CAPE structural antioxidant activity nor to its traditional enzymatic activity in decomposing heme but to a yet to be determined activity. caffeic acid phenethyl ester 62-66 heme oxygenase 1 Homo sapiens 44-48 18312299-4 2008 METHODS: DNA of 99 patients that underwent complete surgical resection of OSCC was analyzed for GTn-repeat polymorphism in the HMOX-1 promoter by polymerase chain reaction, capillary electrophoresis and DNA sequencing. Nitroglycerin 96-99 heme oxygenase 1 Homo sapiens 127-133 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Heme 17-21 heme oxygenase 1 Homo sapiens 101-117 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Heme 17-21 heme oxygenase 1 Homo sapiens 119-123 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Heme 43-47 heme oxygenase 1 Homo sapiens 101-117 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Heme 43-47 heme oxygenase 1 Homo sapiens 119-123 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Biliverdine 62-72 heme oxygenase 1 Homo sapiens 101-117 18657588-2 2008 In the mammalian heme degradation process, heme is cleaved to biliverdin by the rate-limiting enzyme heme oxygenase-1 (HO-1). Biliverdine 62-72 heme oxygenase 1 Homo sapiens 119-123 18459998-1 2008 Heme oxygenase-1 (HO-1) has a microsatellite polymorphism based on the number of guanosine-thymidine nucleotide repeats (GT) repeats that regulates expression levels and could have an impact on organ survival post-injury. guanosine-thymidine nucleotide 81-111 heme oxygenase 1 Homo sapiens 0-16 18459998-1 2008 Heme oxygenase-1 (HO-1) has a microsatellite polymorphism based on the number of guanosine-thymidine nucleotide repeats (GT) repeats that regulates expression levels and could have an impact on organ survival post-injury. guanosine-thymidine nucleotide 81-111 heme oxygenase 1 Homo sapiens 18-22 18584244-0 2008 Evidence that cisplatin-induced auditory damage is attenuated by downregulation of pro-inflammatory cytokines via Nrf2/HO-1. Cisplatin 14-23 heme oxygenase 1 Homo sapiens 119-123 18584244-2 2008 We report here that flunarizine markedly attenuates cisplatin-induced pro-inflammatory cytokine secretion and their messenger RNA transcription as well as cisplatin cytotoxicity through the activation of Nrf2/HO-1 and downregulation of NF-kappaB. Flunarizine 20-31 heme oxygenase 1 Homo sapiens 209-213 18584244-2 2008 We report here that flunarizine markedly attenuates cisplatin-induced pro-inflammatory cytokine secretion and their messenger RNA transcription as well as cisplatin cytotoxicity through the activation of Nrf2/HO-1 and downregulation of NF-kappaB. Cisplatin 52-61 heme oxygenase 1 Homo sapiens 209-213 18584244-3 2008 In HEI-OC1 cells, overexpression of Nrf2/HO-1 by gene transfer or pharmacological approaches attenuated cisplatin-induced cytotoxicity and pro-inflammatory cytokine production. Cisplatin 104-113 heme oxygenase 1 Homo sapiens 41-45 18644421-4 2008 Furthermore, RA stimulated the antioxidant enzyme heme oxygenase-1 (HO-1). rosmarinic acid 13-15 heme oxygenase 1 Homo sapiens 50-66 18644421-4 2008 Furthermore, RA stimulated the antioxidant enzyme heme oxygenase-1 (HO-1). rosmarinic acid 13-15 heme oxygenase 1 Homo sapiens 68-72 18644421-5 2008 We also demonstrated that the HO-1 induction by RA was associated with the protein kinase A (PKA) and phosphatidylinositiol-3-kinase (PI3K) signaling pathways. rosmarinic acid 48-50 heme oxygenase 1 Homo sapiens 30-34 18776993-0 2008 Up-regulation of heme oxygenase-1 by isoflurane preconditioning during tolerance against neuronal injury induced by oxygen glucose deprivation. Isoflurane 37-47 heme oxygenase 1 Homo sapiens 17-33 18776993-2 2008 The HO-1 isozyme is induced by a variety of factors such as heat, heme, ischemia, and hydrogen peroxide. Heme 66-70 heme oxygenase 1 Homo sapiens 4-8 18776993-2 2008 The HO-1 isozyme is induced by a variety of factors such as heat, heme, ischemia, and hydrogen peroxide. Hydrogen Peroxide 86-103 heme oxygenase 1 Homo sapiens 4-8 18776993-5 2008 Recently, isoflurane has been shown to up-regulate HO-1 in the liver. Isoflurane 10-20 heme oxygenase 1 Homo sapiens 51-55 18776993-7 2008 Further study by reverse transcription-polymerase chain reaction and Western blot analysis showed that isoflurane preconditioning significantly increases HO-1 expression in oxygen glucose deprivation (OGD)-induced neuronal injury. Isoflurane 103-113 heme oxygenase 1 Homo sapiens 154-158 18776993-9 2008 These findings indicated that the neuroprotective role of isoflurane preconditioning against OGD-induced injury might be associated with its role in up-regulating HO-1 in ischemic neurons. Isoflurane 58-68 heme oxygenase 1 Homo sapiens 163-167 18584244-4 2008 On the contrary, inhibition of Nrf2/HO-1 signaling by pharmacological inhibitors or specific small interfering RNAs significantly abolished the beneficial effects of flunarizine. Flunarizine 166-177 heme oxygenase 1 Homo sapiens 36-40 18584244-7 2008 Finally, orally administrated Sibelium, the trade name of flunarizine, suppressed the increase of pro-inflammatory cytokines by cisplatin in both serum and cochleas of mice, whereas it increased HO-1 expression in cochleas. Flunarizine 30-38 heme oxygenase 1 Homo sapiens 195-199 18584244-8 2008 These results indicate that flunarizine induces a protective effect against cisplatin ototoxicity through the downregulation of NF-kappaB by Nrf2/HO-1 activation and the resulting inhibition of pro-inflammatory cytokine production in vitro and in vivo. Flunarizine 28-39 heme oxygenase 1 Homo sapiens 146-150 18584244-8 2008 These results indicate that flunarizine induces a protective effect against cisplatin ototoxicity through the downregulation of NF-kappaB by Nrf2/HO-1 activation and the resulting inhibition of pro-inflammatory cytokine production in vitro and in vivo. Cisplatin 76-85 heme oxygenase 1 Homo sapiens 146-150 18631371-6 2008 Additionally, H(2)O(2) treatment induced upregulation of CHOP, GRP78 and several representative endoplasmic reticulum (ER) stress-responsive proteins, including heme oxygenase-1. Hydrogen Peroxide 14-22 heme oxygenase 1 Homo sapiens 161-177 18412543-3 2008 Fluorescence correlation spectroscopy was used to investigate the mobility of hBVR in living cells and its function in the nuclear transport of haematin for induction of HO-1. Hemin 144-152 heme oxygenase 1 Homo sapiens 170-174 18556172-9 2008 Incidentally, MT, Hsp70 and HO-1 showed similar responses to Cd exposure. Cadmium 61-63 heme oxygenase 1 Homo sapiens 28-32 18539158-2 2008 In this study, we found that the expressions of anti-oxidant proteins (gamma-glutamylcysteine ligase heavy chain (gamma-GCL h), heme oxygenase-1, thioredoxin and peroxiredoxin1) in TAM-resistant MCF-7 (TAMR-MCF-7) cells were higher than control MCF-7 cells. Tamoxifen 181-184 heme oxygenase 1 Homo sapiens 128-144 18556172-7 2008 It remained essentially unchanged with 2.5 microM Cd exposure of Sertoli cells; however, MT, Hsp70 and HO-1 were significantly increased by Cd exposure. Cadmium 140-142 heme oxygenase 1 Homo sapiens 103-107 18556172-8 2008 As a result, Cd-induced MT was protected Sertoli cells against apoptosis, and Cd-induced HO-1 was involved in protection against oxidative stress. Cadmium 78-80 heme oxygenase 1 Homo sapiens 89-93 18424257-2 2008 Here, we report that treatment of human breast epithelial (MCF10A) cells with EGCG induces the expression of glutamate-cysteine ligase, manganese superoxide dismutase (MnSOD), and heme oxygenase-1 (HO-1). epigallocatechin gallate 78-82 heme oxygenase 1 Homo sapiens 180-196 18424257-2 2008 Here, we report that treatment of human breast epithelial (MCF10A) cells with EGCG induces the expression of glutamate-cysteine ligase, manganese superoxide dismutase (MnSOD), and heme oxygenase-1 (HO-1). epigallocatechin gallate 78-82 heme oxygenase 1 Homo sapiens 198-202 18424257-5 2008 Silencing of Nrf2 by siRNA gene knockdown rendered the MCF10A cells less sensitive to the EGCG-induced expression of HO-1 and MnSOD. epigallocatechin gallate 90-94 heme oxygenase 1 Homo sapiens 117-121 18550526-0 2008 BACH1 is a specific repressor of HMOX1 that is inactivated by arsenite. arsenite 62-70 heme oxygenase 1 Homo sapiens 33-38 18550526-2 2008 To prevent accumulation, the inducible enzyme heme oxygenase-1 (HMOX1) catalyzes degradation of heme. Heme 46-50 heme oxygenase 1 Homo sapiens 64-69 18550526-4 2008 Conversely, increased intracellular heme or sulfhydryl oxidation inactivate BACH1, permitting transcriptional induction of HMOX1. Heme 36-40 heme oxygenase 1 Homo sapiens 123-128 18550526-6 2008 We show that BACH1 is inactivated at low micromolar arsenite concentrations and that BACH1 inactivation is necessary and sufficient for transcriptional induction of HMOX1. arsenite 52-60 heme oxygenase 1 Homo sapiens 165-170 18550526-8 2008 The loss of BACH1 function in human keratinocytes results almost exclusively in HMOX1 induction, suggesting that BACH1 may function as a rheostat regulating levels of intracellular free heme. Heme 186-190 heme oxygenase 1 Homo sapiens 80-85 18219551-0 2008 Heme oxygenase-1 participates in the anti-inflammatory activity of taurine chloramine. N-chlorotaurine 67-85 heme oxygenase 1 Homo sapiens 0-16 18412543-9 2008 Experiments with nuclear localization, export signal mutants and si-hBVR [siRNA (small interfering RNA) specific to hBVR] indicate that nuclear localization of hBVR is required for induction of HO-1 by haematin. Hemin 202-210 heme oxygenase 1 Homo sapiens 194-198 18443197-0 2008 Serum bilirubin and ferritin levels link heme oxygenase-1 gene promoter polymorphism and susceptibility to coronary artery disease in diabetic patients. Bilirubin 6-15 heme oxygenase 1 Homo sapiens 41-57 18712632-4 2008 The results show that alpha(1)-microglobulin prevents intracellular oxidation and up-regulation of heme oxygenase-1 induced by heme, hydrogen peroxide and Fenton reaction-generated hydroxyl radicals in the culture medium. Hydrogen Peroxide 133-150 heme oxygenase 1 Homo sapiens 99-115 18712632-4 2008 The results show that alpha(1)-microglobulin prevents intracellular oxidation and up-regulation of heme oxygenase-1 induced by heme, hydrogen peroxide and Fenton reaction-generated hydroxyl radicals in the culture medium. Hydroxyl Radical 181-198 heme oxygenase 1 Homo sapiens 99-115 18440590-10 2008 In patients with CAF-MD, the inducible enzyme HO-1 is more expressed in the left atrial areas that show greater structural remodeling. caf-md 17-23 heme oxygenase 1 Homo sapiens 46-50 18443197-3 2008 The aim of this study was to assess the association of the length of (GT)(n) repeats in the HO-1 gene promoter with serum bilirubin, markers of iron status, and the development of coronary artery disease (CAD). Bilirubin 122-131 heme oxygenase 1 Homo sapiens 92-96 18443197-3 2008 The aim of this study was to assess the association of the length of (GT)(n) repeats in the HO-1 gene promoter with serum bilirubin, markers of iron status, and the development of coronary artery disease (CAD). Iron 144-148 heme oxygenase 1 Homo sapiens 92-96 18443197-9 2008 With adjustment for both serum bilirubin and ferritin, the effect of HO-1 promoter polymorphism on susceptibility to CAD disappeared. Bilirubin 31-40 heme oxygenase 1 Homo sapiens 69-73 18443197-10 2008 CONCLUSIONS: Length polymorphism in the HO-1 gene promoter is correlated with susceptibility to CAD in diabetic patients, and this effect might be conveyed through its influence on serum bilirubin and ferritin. Bilirubin 187-196 heme oxygenase 1 Homo sapiens 40-44 18331200-1 2008 Heme oxygenase-1 is the rate-limiting enzyme for the degradation of the prooxidant heme. Heme 83-87 heme oxygenase 1 Homo sapiens 0-16 18634931-0 2008 Hydrogen peroxide induces heme oxygenase-1 and dentin sialophosphoprotein mRNA in human pulp cells. Hydrogen Peroxide 0-17 heme oxygenase 1 Homo sapiens 26-42 18634931-1 2008 Although the induction of heme oxygenase-1 (HO-1) by hydrogen peroxide (H2O2) has been reported, the HO-1 and odontoblastic differentiation-inducing effects against H2O2 have not been clarified in human pulp cells. Hydrogen Peroxide 53-70 heme oxygenase 1 Homo sapiens 26-42 18634931-1 2008 Although the induction of heme oxygenase-1 (HO-1) by hydrogen peroxide (H2O2) has been reported, the HO-1 and odontoblastic differentiation-inducing effects against H2O2 have not been clarified in human pulp cells. Hydrogen Peroxide 53-70 heme oxygenase 1 Homo sapiens 44-48 18634931-1 2008 Although the induction of heme oxygenase-1 (HO-1) by hydrogen peroxide (H2O2) has been reported, the HO-1 and odontoblastic differentiation-inducing effects against H2O2 have not been clarified in human pulp cells. Hydrogen Peroxide 72-76 heme oxygenase 1 Homo sapiens 26-42 18634931-1 2008 Although the induction of heme oxygenase-1 (HO-1) by hydrogen peroxide (H2O2) has been reported, the HO-1 and odontoblastic differentiation-inducing effects against H2O2 have not been clarified in human pulp cells. Hydrogen Peroxide 72-76 heme oxygenase 1 Homo sapiens 44-48 18634931-3 2008 H2O2 decreased cell viability but increased HO-1 and DSPP expression in a concentration- and time-dependent manner. Hydrogen Peroxide 0-4 heme oxygenase 1 Homo sapiens 44-48 18634931-4 2008 Antioxidants and inhibitors of HO-1, phosphatidylinositol-3"-kinase, extracellular signal-regulated kinase, and p38 mitogen-activated protein kinase blocked H2O2-induced cytotoxicity and the expression of HO-1 and DSPP mRNA in pulp cells. Hydrogen Peroxide 157-161 heme oxygenase 1 Homo sapiens 31-35 18634931-4 2008 Antioxidants and inhibitors of HO-1, phosphatidylinositol-3"-kinase, extracellular signal-regulated kinase, and p38 mitogen-activated protein kinase blocked H2O2-induced cytotoxicity and the expression of HO-1 and DSPP mRNA in pulp cells. Hydrogen Peroxide 157-161 heme oxygenase 1 Homo sapiens 205-209 18634931-5 2008 These data suggest that the induction of HO-1 by H2O2 in pulp cells plays a protective role against the cytotoxic effects of H2O2 and stimulates DSPP expression, resulting in premature odontoblast differentiation through pathways that involve phosphatidylinositol-3"-kinase, p38, and extracellular signal-regulated kinase. Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 41-45 18634931-5 2008 These data suggest that the induction of HO-1 by H2O2 in pulp cells plays a protective role against the cytotoxic effects of H2O2 and stimulates DSPP expression, resulting in premature odontoblast differentiation through pathways that involve phosphatidylinositol-3"-kinase, p38, and extracellular signal-regulated kinase. Hydrogen Peroxide 125-129 heme oxygenase 1 Homo sapiens 41-45 18636201-5 2008 In addition, combination treatment with As2O3 and sulindac increased reactive oxygen species (ROS) and oxidative stress, as evidenced by the heme oxygenase-1 (HO-1) expression and mitogen-activated protein kinase (MAPK) phosphorylation. Arsenic Trioxide 40-45 heme oxygenase 1 Homo sapiens 141-157 18636201-5 2008 In addition, combination treatment with As2O3 and sulindac increased reactive oxygen species (ROS) and oxidative stress, as evidenced by the heme oxygenase-1 (HO-1) expression and mitogen-activated protein kinase (MAPK) phosphorylation. Arsenic Trioxide 40-45 heme oxygenase 1 Homo sapiens 159-163 18636201-5 2008 In addition, combination treatment with As2O3 and sulindac increased reactive oxygen species (ROS) and oxidative stress, as evidenced by the heme oxygenase-1 (HO-1) expression and mitogen-activated protein kinase (MAPK) phosphorylation. Sulindac 50-58 heme oxygenase 1 Homo sapiens 141-157 18636201-5 2008 In addition, combination treatment with As2O3 and sulindac increased reactive oxygen species (ROS) and oxidative stress, as evidenced by the heme oxygenase-1 (HO-1) expression and mitogen-activated protein kinase (MAPK) phosphorylation. Sulindac 50-58 heme oxygenase 1 Homo sapiens 159-163 18636201-8 2008 Combination treatment with As2O3 and sulindac induced oxidative DNA damage in a time-dependent fashion, which was evaluated by H2AX phosphorylation along with HO-1 induction. Arsenic Trioxide 27-32 heme oxygenase 1 Homo sapiens 159-163 18636201-8 2008 Combination treatment with As2O3 and sulindac induced oxidative DNA damage in a time-dependent fashion, which was evaluated by H2AX phosphorylation along with HO-1 induction. Sulindac 37-45 heme oxygenase 1 Homo sapiens 159-163 17964723-6 2008 Human embryonic kidney (HEK) 293T cells treated with 10 microM CoPP expressed 20-fold higher HO-1 levels when compared to controls; this effect was suppressed by transfection with the dominant negative for the nuclear factor-erythroid 2-related factor 2 (Nrf2). cobaltiprotoporphyrin 63-67 heme oxygenase 1 Homo sapiens 93-97 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Oxygen 45-47 heme oxygenase 1 Homo sapiens 6-22 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Oxygen 45-47 heme oxygenase 1 Homo sapiens 24-29 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. NADP 53-58 heme oxygenase 1 Homo sapiens 6-22 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. NADP 53-58 heme oxygenase 1 Homo sapiens 24-29 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Heme 6-10 heme oxygenase 1 Homo sapiens 24-29 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 90-100 heme oxygenase 1 Homo sapiens 6-22 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 90-100 heme oxygenase 1 Homo sapiens 24-29 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 102-104 heme oxygenase 1 Homo sapiens 6-22 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 102-104 heme oxygenase 1 Homo sapiens 24-29 18593583-0 2008 The coffee diterpene kahweol induces heme oxygenase-1 via the PI3K and p38/Nrf2 pathway to protect human dopaminergic neurons from 6-hydroxydopamine-derived oxidative stress. Diterpenes 11-20 heme oxygenase 1 Homo sapiens 37-53 18593583-0 2008 The coffee diterpene kahweol induces heme oxygenase-1 via the PI3K and p38/Nrf2 pathway to protect human dopaminergic neurons from 6-hydroxydopamine-derived oxidative stress. kahweol 21-28 heme oxygenase 1 Homo sapiens 37-53 18593583-0 2008 The coffee diterpene kahweol induces heme oxygenase-1 via the PI3K and p38/Nrf2 pathway to protect human dopaminergic neurons from 6-hydroxydopamine-derived oxidative stress. Oxidopamine 131-148 heme oxygenase 1 Homo sapiens 37-53 18593583-3 2008 Kahweol also up-regulated heme oxygenase-1 (HO-1) expression, which conferred neuroprotection against 6-OHDA-induced oxidative injury. Oxidopamine 102-108 heme oxygenase 1 Homo sapiens 26-42 18593583-3 2008 Kahweol also up-regulated heme oxygenase-1 (HO-1) expression, which conferred neuroprotection against 6-OHDA-induced oxidative injury. Oxidopamine 102-108 heme oxygenase 1 Homo sapiens 44-48 18593583-4 2008 Moreover, kahweol induced PI3K and p38 activation, which are involved in the induction of Nrf2, HO-1 expression, and neuroprotection. kahweol 10-17 heme oxygenase 1 Homo sapiens 96-100 18593583-5 2008 These results suggest that regulation of the anti-oxidant enzyme HO-1 via the PI3K and p38/Nrf2 signaling pathways controls the intracellular levels of ROS. ros 152-155 heme oxygenase 1 Homo sapiens 65-69 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 6-22 18487208-2 2008 Human heme oxygenase-1 (hHO-1) catalyzes the O2- and NADPH-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 115-119 heme oxygenase 1 Homo sapiens 24-29 18487208-4 2008 Here we report spectroscopic detection of a transient intermediate during oxidation by hHO-1 of alpha-meso-phenylheme-IX, alpha-meso-(p-methylphenyl)-mesoheme-III, and alpha-meso-(p-trifluoromethylphenyl)-mesoheme-III. alpha-meso-phenylheme-ix 96-120 heme oxygenase 1 Homo sapiens 87-92 18487208-4 2008 Here we report spectroscopic detection of a transient intermediate during oxidation by hHO-1 of alpha-meso-phenylheme-IX, alpha-meso-(p-methylphenyl)-mesoheme-III, and alpha-meso-(p-trifluoromethylphenyl)-mesoheme-III. alpha-meso-(p-methylphenyl)-mesoheme 122-158 heme oxygenase 1 Homo sapiens 87-92 18487208-4 2008 Here we report spectroscopic detection of a transient intermediate during oxidation by hHO-1 of alpha-meso-phenylheme-IX, alpha-meso-(p-methylphenyl)-mesoheme-III, and alpha-meso-(p-trifluoromethylphenyl)-mesoheme-III. alpha-meso-(p-trifluoromethylphenyl)-mesoheme 168-213 heme oxygenase 1 Homo sapiens 87-92 18487208-11 2008 The hHO-1-isoporphyrin complexes were found to have half-lives of 1.7 and 2.4 h for the p-trifluoromethyl- and p-methyl-substituted phenylhemes, respectively. p-trifluoromethyl- and p-methyl-substituted phenylhemes 88-143 heme oxygenase 1 Homo sapiens 4-9 18487208-12 2008 The addition of NADPH-P450 reductase to the H2O2-generated hHO-1-isoporphyrin complex produced alpha-biliverdin, confirming its role as a reaction intermediate. Hydrogen Peroxide 44-48 heme oxygenase 1 Homo sapiens 59-64 18487208-12 2008 The addition of NADPH-P450 reductase to the H2O2-generated hHO-1-isoporphyrin complex produced alpha-biliverdin, confirming its role as a reaction intermediate. biliverdin 95-111 heme oxygenase 1 Homo sapiens 59-64 18487208-13 2008 Identification of an isoporphyrin intermediate in the catalytic sequence of hHO-1, the first such intermediate observed in hemoprotein catalysis, completes our understanding of the critical first step of heme oxidation. isoporphyrin 21-33 heme oxygenase 1 Homo sapiens 76-81 18487208-13 2008 Identification of an isoporphyrin intermediate in the catalytic sequence of hHO-1, the first such intermediate observed in hemoprotein catalysis, completes our understanding of the critical first step of heme oxidation. Heme 204-208 heme oxygenase 1 Homo sapiens 76-81 18435907-5 2008 Interestingly, the effects of tranilast on LPS-induced PGE(2), NO, TNF-alpha, and IL-1beta production were partially reversed by the HO-1 inhibitor tin protoporphyrin, suggesting that tranilast-induced HO-1 expression is at least partly responsible for the resulting anti-inflammatory effects of the drug. protoporphyrin IX 152-166 heme oxygenase 1 Homo sapiens 133-137 18435907-5 2008 Interestingly, the effects of tranilast on LPS-induced PGE(2), NO, TNF-alpha, and IL-1beta production were partially reversed by the HO-1 inhibitor tin protoporphyrin, suggesting that tranilast-induced HO-1 expression is at least partly responsible for the resulting anti-inflammatory effects of the drug. protoporphyrin IX 152-166 heme oxygenase 1 Homo sapiens 202-206 18452993-5 2008 In addition, L-theanine partially prevented both rotenone- and dieldrin-induced heme oxygenase-1 (HO-1) up-regulation. theanine 13-23 heme oxygenase 1 Homo sapiens 80-96 18547064-0 2008 Structure-activity relationships of methoxychalcones as inducers of heme oxygenase-1. methoxychalcones 36-52 heme oxygenase 1 Homo sapiens 68-84 18547064-3 2008 Recent evidence indicates that the bioactivity of hydroxy-chalcones is correlated with their intrinsic property to induce the antioxidant and cytoprotective enzyme heme oxygenase-1 (HO-1). hydroxy-chalcones 50-67 heme oxygenase 1 Homo sapiens 164-180 18547064-3 2008 Recent evidence indicates that the bioactivity of hydroxy-chalcones is correlated with their intrinsic property to induce the antioxidant and cytoprotective enzyme heme oxygenase-1 (HO-1). hydroxy-chalcones 50-67 heme oxygenase 1 Homo sapiens 182-186 18547064-7 2008 This study provides additional information on the structural features that methoxychalcones and natural antioxidants need to possess to be considered as therapeutic agents for maximizing HO-1 expression and activity. methoxychalcones 75-91 heme oxygenase 1 Homo sapiens 187-191 18691515-7 2008 Dermal fibroblasts exposed to 25 micromol/L ferulic acid ethyl ester in the presence of 500 micromol/L hydrogen peroxide showed an increased level of both heme oxygenase-1 and heat shock protein-70 compared with dermal fibroblasts treated with hydrogen peroxide alone. ethyl ferulate 44-68 heme oxygenase 1 Homo sapiens 155-197 18691515-7 2008 Dermal fibroblasts exposed to 25 micromol/L ferulic acid ethyl ester in the presence of 500 micromol/L hydrogen peroxide showed an increased level of both heme oxygenase-1 and heat shock protein-70 compared with dermal fibroblasts treated with hydrogen peroxide alone. Hydrogen Peroxide 103-120 heme oxygenase 1 Homo sapiens 155-197 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Heme 22-26 heme oxygenase 1 Homo sapiens 40-44 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Carbon Monoxide 81-96 heme oxygenase 1 Homo sapiens 22-38 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Carbon Monoxide 81-96 heme oxygenase 1 Homo sapiens 40-44 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Carbon Monoxide 98-100 heme oxygenase 1 Homo sapiens 22-38 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Carbon Monoxide 98-100 heme oxygenase 1 Homo sapiens 40-44 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 22-38 18381758-1 2008 The inducible protein heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to carbon monoxide (CO) and biliverdin, which play a concerted action in cytoprotection against oxidative stress and in the modulation of cell proliferation and differentiation. Biliverdine 106-116 heme oxygenase 1 Homo sapiens 40-44 18381758-2 2008 Here we report that both HO-1 expression and activity can be highly increased in undifferentiated human mesenchymal stem cells (MSCs) treated with hemin, a known HO-1 inducer. Hemin 147-152 heme oxygenase 1 Homo sapiens 25-29 18381758-2 2008 Here we report that both HO-1 expression and activity can be highly increased in undifferentiated human mesenchymal stem cells (MSCs) treated with hemin, a known HO-1 inducer. Hemin 147-152 heme oxygenase 1 Homo sapiens 162-166 18381758-3 2008 However, HO-1 mRNA and protein expression gradually decrease when MSCs undergo neural differentiation in vitro, making them extremely susceptible to glutamate-mediated cytotoxicity. Glutamic Acid 149-158 heme oxygenase 1 Homo sapiens 9-13 18460015-2 2008 A HO-1 gene promoter microsatellite (GT)(n) dinucleotide repeat polymorphism was associated with regulation of HO-1 in response to inflammatory stimuli. (n) dinucleotide 40-56 heme oxygenase 1 Homo sapiens 2-6 18460015-2 2008 A HO-1 gene promoter microsatellite (GT)(n) dinucleotide repeat polymorphism was associated with regulation of HO-1 in response to inflammatory stimuli. (n) dinucleotide 40-56 heme oxygenase 1 Homo sapiens 111-115 18452993-5 2008 In addition, L-theanine partially prevented both rotenone- and dieldrin-induced heme oxygenase-1 (HO-1) up-regulation. Rotenone 49-57 heme oxygenase 1 Homo sapiens 80-96 18452993-5 2008 In addition, L-theanine partially prevented both rotenone- and dieldrin-induced heme oxygenase-1 (HO-1) up-regulation. Dieldrin 63-71 heme oxygenase 1 Homo sapiens 80-96 17712531-8 2008 Consistent with this, when the acidification of endosomes is prevented by bafilomycin then heme oxygenase-1 induction by heme-HPX no longer occurs. bafilomycin 74-85 heme oxygenase 1 Homo sapiens 91-107 18567745-0 2008 Enterolactone induces heme oxygenase-1 expression through nuclear factor-E2-related factor 2 activation in endothelial cells. 2,3-bis(3'-hydroxybenzyl)butyrolactone 0-13 heme oxygenase 1 Homo sapiens 22-38 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Bilirubin 202-211 heme oxygenase 1 Homo sapiens 109-125 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Bilirubin 202-211 heme oxygenase 1 Homo sapiens 127-131 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Biliverdine 216-226 heme oxygenase 1 Homo sapiens 109-125 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Biliverdine 216-226 heme oxygenase 1 Homo sapiens 127-131 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Carbon Monoxide 238-253 heme oxygenase 1 Homo sapiens 109-125 18567745-3 2008 Moreover, many, but not all, phenolic compounds can have indirect antioxidative effects through induction of heme oxygenase-1 (HO-1), which has antiinflammatory functions via production of antioxidants bilirubin and biliverdin as well as carbon monoxide, thereby contributing to cardiovascular health. Carbon Monoxide 238-253 heme oxygenase 1 Homo sapiens 127-131 18567745-4 2008 Our aim was therefore to assess whether enterolactone has indirect antioxidative effects via induction of HO-1 in endothelial cells. 2,3-bis(3'-hydroxybenzyl)butyrolactone 40-53 heme oxygenase 1 Homo sapiens 106-110 18567745-5 2008 The effect of enterolactone on HO-1 mRNA and protein expression in human umbilical vein endothelial cells (HUVEC) was analyzed by quantitative real-time PCR and western blot. 2,3-bis(3'-hydroxybenzyl)butyrolactone 14-27 heme oxygenase 1 Homo sapiens 31-35 18567745-6 2008 The role of nuclear factor-E2-related factor 2 (Nrf2) in HO-1 induction by enterolactone was studied using small interfering RNA (siRNA) and chromatin immunoprecipitation (ChIP) methods. 2,3-bis(3'-hydroxybenzyl)butyrolactone 75-88 heme oxygenase 1 Homo sapiens 57-61 18567745-7 2008 Our results showed that enterolactone induced HO-1 in HUVEC in a time- and concentration-dependent manner. 2,3-bis(3'-hydroxybenzyl)butyrolactone 24-37 heme oxygenase 1 Homo sapiens 46-50 18567745-8 2008 The induction appeared to be mediated via the transcription factor Nrf2, as Nrf2 siRNA abolished the HO-1 induction by enterolactone. 2,3-bis(3'-hydroxybenzyl)butyrolactone 119-132 heme oxygenase 1 Homo sapiens 101-105 18567745-9 2008 We also showed using ChIP that exposure to enterolactone increased the binding of Nrf2 to the promoter region of HO-1. 2,3-bis(3'-hydroxybenzyl)butyrolactone 43-56 heme oxygenase 1 Homo sapiens 113-117 18567745-10 2008 In conclusion, enterolactone increases the expression of HO-1 via Nrf2, which may contribute to its vasculoprotective effects. 2,3-bis(3'-hydroxybenzyl)butyrolactone 15-28 heme oxygenase 1 Homo sapiens 57-61 18440740-9 2008 We also found that PD98059, a MEK (MAPK/ERK kinase) inhibitor, attenuated the eupatilin-induced HO-1 expression and nuclear translocation of transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2). eupatilin 78-87 heme oxygenase 1 Homo sapiens 96-100 18523255-1 2008 The induction of heme oxygenase 1 (HO-1) by a single treatment with cobalt protoporphyrin (CoPPIX) protects against inflammatory liver failure and ischemia reperfusion injury after allotransplantation. cobaltiprotoporphyrin 68-89 heme oxygenase 1 Homo sapiens 17-33 18523255-1 2008 The induction of heme oxygenase 1 (HO-1) by a single treatment with cobalt protoporphyrin (CoPPIX) protects against inflammatory liver failure and ischemia reperfusion injury after allotransplantation. cobaltiprotoporphyrin 68-89 heme oxygenase 1 Homo sapiens 35-39 18523255-1 2008 The induction of heme oxygenase 1 (HO-1) by a single treatment with cobalt protoporphyrin (CoPPIX) protects against inflammatory liver failure and ischemia reperfusion injury after allotransplantation. coppix 91-97 heme oxygenase 1 Homo sapiens 17-33 18523255-1 2008 The induction of heme oxygenase 1 (HO-1) by a single treatment with cobalt protoporphyrin (CoPPIX) protects against inflammatory liver failure and ischemia reperfusion injury after allotransplantation. coppix 91-97 heme oxygenase 1 Homo sapiens 35-39 18523255-3 2008 To investigate the poorly understood mechanism of CoPPIX-induced HO-1 activity in more detail, we performed gene expression analysis in murine liver, revealing the up-regulation of STAT3 after CoPPIX treatment. coppix 50-56 heme oxygenase 1 Homo sapiens 65-69 18523255-3 2008 To investigate the poorly understood mechanism of CoPPIX-induced HO-1 activity in more detail, we performed gene expression analysis in murine liver, revealing the up-regulation of STAT3 after CoPPIX treatment. coppix 193-199 heme oxygenase 1 Homo sapiens 65-69 18598766-6 2008 PI3K inhibitor LY294002 could block the elevation of HO-1 protein expression and reverse the protective effect of alpha-Gal silencing and GS-IB4 against CDC. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 15-23 heme oxygenase 1 Homo sapiens 53-57 18440740-7 2008 Heme oxygenase-1 (HO-1), which is known as a cytoprotective enzyme due to its anti-inflammatory actions, is a candidate protein since ZnPP, an HO-1 inhibitor, repressed the protective effect of eupatilin on indomethacin-induced cell damage in a concentration-dependent manner. zinc protoporphyrin 134-138 heme oxygenase 1 Homo sapiens 0-16 18440740-7 2008 Heme oxygenase-1 (HO-1), which is known as a cytoprotective enzyme due to its anti-inflammatory actions, is a candidate protein since ZnPP, an HO-1 inhibitor, repressed the protective effect of eupatilin on indomethacin-induced cell damage in a concentration-dependent manner. zinc protoporphyrin 134-138 heme oxygenase 1 Homo sapiens 18-22 18440740-7 2008 Heme oxygenase-1 (HO-1), which is known as a cytoprotective enzyme due to its anti-inflammatory actions, is a candidate protein since ZnPP, an HO-1 inhibitor, repressed the protective effect of eupatilin on indomethacin-induced cell damage in a concentration-dependent manner. zinc protoporphyrin 134-138 heme oxygenase 1 Homo sapiens 143-147 18440740-7 2008 Heme oxygenase-1 (HO-1), which is known as a cytoprotective enzyme due to its anti-inflammatory actions, is a candidate protein since ZnPP, an HO-1 inhibitor, repressed the protective effect of eupatilin on indomethacin-induced cell damage in a concentration-dependent manner. Indomethacin 207-219 heme oxygenase 1 Homo sapiens 0-16 18440740-7 2008 Heme oxygenase-1 (HO-1), which is known as a cytoprotective enzyme due to its anti-inflammatory actions, is a candidate protein since ZnPP, an HO-1 inhibitor, repressed the protective effect of eupatilin on indomethacin-induced cell damage in a concentration-dependent manner. Indomethacin 207-219 heme oxygenase 1 Homo sapiens 18-22 18440740-9 2008 We also found that PD98059, a MEK (MAPK/ERK kinase) inhibitor, attenuated the eupatilin-induced HO-1 expression and nuclear translocation of transcription factor Nrf2 (nuclear factor erythroid 2-related factor 2). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 19-26 heme oxygenase 1 Homo sapiens 96-100 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. S-Nitroso-N-propionyl-D,L-penicillamine 138-142 heme oxygenase 1 Homo sapiens 80-84 18598163-0 2008 Luteolin suppresses cisplatin-induced apoptosis in auditory cells: possible mediation through induction of heme oxygenase-1 expression. Cisplatin 20-29 heme oxygenase 1 Homo sapiens 107-123 18598163-8 2008 These results demonstrate that the expression of HO-1 by luteolin is mediated by the ERK pathway, and also that the activating of HO-1 inhibits cisplatin-induced apoptosis in HEI-OC1 1 cells. Cisplatin 144-153 heme oxygenase 1 Homo sapiens 49-53 18598163-8 2008 These results demonstrate that the expression of HO-1 by luteolin is mediated by the ERK pathway, and also that the activating of HO-1 inhibits cisplatin-induced apoptosis in HEI-OC1 1 cells. Cisplatin 144-153 heme oxygenase 1 Homo sapiens 130-134 18357586-0 2008 Heme oxygenase-1 mediates the anti-inflammatory effect of Curcumin within LPS-stimulated human monocytes. Curcumin 58-66 heme oxygenase 1 Homo sapiens 0-16 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. Curcumin 0-8 heme oxygenase 1 Homo sapiens 80-84 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. Curcumin 0-8 heme oxygenase 1 Homo sapiens 196-200 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. polyphenolic compound 12-33 heme oxygenase 1 Homo sapiens 80-84 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. polyphenolic compound 12-33 heme oxygenase 1 Homo sapiens 196-200 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. Curcumin 137-145 heme oxygenase 1 Homo sapiens 80-84 18357586-1 2008 Curcumin, a polyphenolic compound derived from plant, regulates heme oxygenase (HO-1) expression within certain cell types; however, the Curcumin-mediated signal transduction in the regulation of HO-1 expression within human monocytes/macrophages is unclear. Curcumin 137-145 heme oxygenase 1 Homo sapiens 196-200 18357586-2 2008 Herein, we show that Curcumin dose dependently induced HO-1 expression and HO-1 activity through the activation of PKCalpha, PKCdelta/ERK1/2, p38alpha, and PI3-kinase. Curcumin 21-29 heme oxygenase 1 Homo sapiens 55-59 18357586-2 2008 Herein, we show that Curcumin dose dependently induced HO-1 expression and HO-1 activity through the activation of PKCalpha, PKCdelta/ERK1/2, p38alpha, and PI3-kinase. Curcumin 21-29 heme oxygenase 1 Homo sapiens 75-79 18357586-3 2008 In addition, H2O2 release is essential for Curcumin-mediated ERK1/2 and p38 phosphorylation and HO-1 expression. Hydrogen Peroxide 13-17 heme oxygenase 1 Homo sapiens 96-100 18357586-3 2008 In addition, H2O2 release is essential for Curcumin-mediated ERK1/2 and p38 phosphorylation and HO-1 expression. Curcumin 43-51 heme oxygenase 1 Homo sapiens 96-100 18498885-0 2008 Effect of mineral trioxide aggregate on dentin bridge formation and expression of dentin sialoprotein and heme oxygenase-1 in human dental pulp. mineral trioxide 10-26 heme oxygenase 1 Homo sapiens 106-122 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. S-Nitroso-N-propionyl-D,L-penicillamine 138-142 heme oxygenase 1 Homo sapiens 108-112 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. S-Nitroso-N-propionyl-D,L-penicillamine 138-142 heme oxygenase 1 Homo sapiens 108-112 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. Curcumin 178-186 heme oxygenase 1 Homo sapiens 80-84 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. Curcumin 178-186 heme oxygenase 1 Homo sapiens 108-112 18357586-4 2008 Further, Curcumin inhibited LPS-induced IL-1 and IL-6 secretion and blockage of HO-1 expression/activity by HO-1 siRNA or HO-1 inhibitor, SnPP reversed the inhibitory effects of Curcumin on cytokines secretion. Curcumin 178-186 heme oxygenase 1 Homo sapiens 108-112 18357586-5 2008 HO-1 over-expression produced the same inhibitory effects of Curcumin on IL-1 secretion. Curcumin 61-69 heme oxygenase 1 Homo sapiens 0-4 18357586-6 2008 Collectively, our results suggest that Curcumin inhibits cytokines secretion within LPS-stimulated monocytes through a mechanism that involves the action of HO-1. Curcumin 39-47 heme oxygenase 1 Homo sapiens 157-161 18332044-0 2008 Heme-oxygenase 1 gene expression is a marker for hexavalent chromium-induced stress and toxicity in human dermal fibroblasts. Chromium 60-68 heme oxygenase 1 Homo sapiens 0-16 18332044-5 2008 The Cr(VI)-induced overexpression of heme-oxygenase 1 messenger RNA (HO-1) in the fibroblasts was significantly blocked by actinomycin D and by inhibitors of MAP kinase pathways. Dactinomycin 123-136 heme oxygenase 1 Homo sapiens 37-53 18332044-5 2008 The Cr(VI)-induced overexpression of heme-oxygenase 1 messenger RNA (HO-1) in the fibroblasts was significantly blocked by actinomycin D and by inhibitors of MAP kinase pathways. Dactinomycin 123-136 heme oxygenase 1 Homo sapiens 69-73 18332044-6 2008 The Cr(VI)-induced cytotoxicity and the overexpression of the HO-1 gene were dependent on the glutathione level of the fibroblasts. Glutathione 94-105 heme oxygenase 1 Homo sapiens 62-66 18332044-7 2008 Buthionine sulfoximine-mediated GSH depletion resulted in enhanced Cr(VI) cytotoxicity and further overexpression of the HO-1 gene. Buthionine Sulfoximine 0-22 heme oxygenase 1 Homo sapiens 121-125 18332044-7 2008 Buthionine sulfoximine-mediated GSH depletion resulted in enhanced Cr(VI) cytotoxicity and further overexpression of the HO-1 gene. Glutathione 32-35 heme oxygenase 1 Homo sapiens 121-125 18332044-8 2008 On the other hand, elevated cellular levels of glutathione resulting from pretreating the cells with GSH significantly protected the cells against the Cr(VI)-induced cytotoxicity and blocked the HO-1 gene"s overexpression. Glutathione 47-58 heme oxygenase 1 Homo sapiens 195-199 18332044-8 2008 On the other hand, elevated cellular levels of glutathione resulting from pretreating the cells with GSH significantly protected the cells against the Cr(VI)-induced cytotoxicity and blocked the HO-1 gene"s overexpression. Glutathione 101-104 heme oxygenase 1 Homo sapiens 195-199 18332044-9 2008 Pretreating the fibroblasts with N-acetyl cysteine also significantly reduced the Cr(VI)-induced cytotoxicity and overexpression of the HO-1 gene. Acetylcysteine 33-50 heme oxygenase 1 Homo sapiens 136-140 18332044-9 2008 Pretreating the fibroblasts with N-acetyl cysteine also significantly reduced the Cr(VI)-induced cytotoxicity and overexpression of the HO-1 gene. chromium hexavalent ion 82-88 heme oxygenase 1 Homo sapiens 136-140 18376820-0 2008 Selenolate complexes of CYP101 and the heme-bound hHO-1/H25A proximal cavity mutant. selenolate 0-10 heme oxygenase 1 Homo sapiens 50-55 18172853-6 2008 Protein levels of gamma-GCS and heme oxygenase-1 (HO-1), two MARE-containing genes, were also increased after hemin treatment. Hemin 110-115 heme oxygenase 1 Homo sapiens 32-48 18376820-0 2008 Selenolate complexes of CYP101 and the heme-bound hHO-1/H25A proximal cavity mutant. Heme 39-43 heme oxygenase 1 Homo sapiens 50-55 18376820-1 2008 Thiolate and selenolate complexes of CYP101 (P450cam) and the H25A proximal cavity mutant of heme-bound human heme oxygenase-1 (hHO-1) have been examined by UV-vis spectroscopy. thiolate 0-8 heme oxygenase 1 Homo sapiens 110-126 18376820-1 2008 Thiolate and selenolate complexes of CYP101 (P450cam) and the H25A proximal cavity mutant of heme-bound human heme oxygenase-1 (hHO-1) have been examined by UV-vis spectroscopy. thiolate 0-8 heme oxygenase 1 Homo sapiens 128-133 18376820-1 2008 Thiolate and selenolate complexes of CYP101 (P450cam) and the H25A proximal cavity mutant of heme-bound human heme oxygenase-1 (hHO-1) have been examined by UV-vis spectroscopy. selenolate 13-23 heme oxygenase 1 Homo sapiens 110-126 18376820-1 2008 Thiolate and selenolate complexes of CYP101 (P450cam) and the H25A proximal cavity mutant of heme-bound human heme oxygenase-1 (hHO-1) have been examined by UV-vis spectroscopy. Heme 93-97 heme oxygenase 1 Homo sapiens 110-126 18376820-1 2008 Thiolate and selenolate complexes of CYP101 (P450cam) and the H25A proximal cavity mutant of heme-bound human heme oxygenase-1 (hHO-1) have been examined by UV-vis spectroscopy. Heme 93-97 heme oxygenase 1 Homo sapiens 128-133 18376820-3 2008 Thiolate ligands also bound to the proximal side of the heme in the cavity created by the H25A mutation in hHO-1, giving a Soret absorption similar to that of the H25C hHO-1 mutant. thiolate 0-8 heme oxygenase 1 Homo sapiens 107-112 18376820-3 2008 Thiolate ligands also bound to the proximal side of the heme in the cavity created by the H25A mutation in hHO-1, giving a Soret absorption similar to that of the H25C hHO-1 mutant. thiolate 0-8 heme oxygenase 1 Homo sapiens 168-173 18376820-3 2008 Thiolate ligands also bound to the proximal side of the heme in the cavity created by the H25A mutation in hHO-1, giving a Soret absorption similar to that of the H25C hHO-1 mutant. Heme 56-60 heme oxygenase 1 Homo sapiens 107-112 18376820-3 2008 Thiolate ligands also bound to the proximal side of the heme in the cavity created by the H25A mutation in hHO-1, giving a Soret absorption similar to that of the H25C hHO-1 mutant. Heme 56-60 heme oxygenase 1 Homo sapiens 168-173 18344023-8 2008 Both cobalt-protoporphyrin (CoPP) and CoCl2 markedly induced HO-1 expression. cobaltiprotoporphyrin 5-26 heme oxygenase 1 Homo sapiens 61-65 18492619-11 2008 In addition, arecoline was found to elevate HO-1 mRNA in a dose-dependent manner (p < 0.05). Arecoline 13-22 heme oxygenase 1 Homo sapiens 44-48 18492619-12 2008 The addition of BaP enhanced arecolineinduced HO-1 expression (p < 0.05). Benzo(a)pyrene 16-19 heme oxygenase 1 Homo sapiens 46-50 18492619-13 2008 Moreover, addition of NAC markedly inhibited arecoline-induced HO-1 expression (p < 0.05). Arecoline 45-54 heme oxygenase 1 Homo sapiens 63-67 18492619-14 2008 CONCLUSION: Taken together, these results suggest that HO-1 expression is significantly upregulated in OSCC from areca quid chewers, and arecoline may be responsible for enhanced HO-1 expression in vivo. Arecoline 137-146 heme oxygenase 1 Homo sapiens 179-183 18492619-16 2008 The regulation of HO-1 expression induced by arecoline is critically dependent on intracellular GSH concentration. Arecoline 45-54 heme oxygenase 1 Homo sapiens 18-22 18492619-16 2008 The regulation of HO-1 expression induced by arecoline is critically dependent on intracellular GSH concentration. Glutathione 96-99 heme oxygenase 1 Homo sapiens 18-22 18490853-8 2008 Antisense HO-1 and HSP 72 oligodeoxynucleotide inhibited HO-1 and HSP 72 induction, respectively, and significantly aggravated cerebral vasospasm. Oligodeoxyribonucleotides 26-46 heme oxygenase 1 Homo sapiens 57-61 18227147-5 2008 Moreover, glutathione-depletion and cytochrome P450 (P450)-inhibition experiments have shown that the observed HO-1 induction was triggered by the electrophilic reactive metabolites produced from the problematic drugs through P450-mediated metabolic bioactivation. Glutathione 10-21 heme oxygenase 1 Homo sapiens 111-115 18545641-4 2008 The mechanism of protective actions of HO-1 has not been completely elucidated, but recent evidence suggests that one or more of heme metabolites can mediate the protective effects of HO-1. Heme 129-133 heme oxygenase 1 Homo sapiens 184-188 18205746-0 2008 Differential induction of heme oxygenase-1 against nicotine-induced cytotoxicity via the PI3K, MAPK, and NF-kappa B pathways in immortalized and malignant human oral keratinocytes. Nicotine 51-59 heme oxygenase 1 Homo sapiens 26-42 18205746-1 2008 BACKGROUND: Heme oxygenase-1 (HO-1) exhibits cytoprotective effects in many different cell types and is induced by nicotine exposure in human gingival fibroblasts. Nicotine 115-123 heme oxygenase 1 Homo sapiens 12-28 18205746-1 2008 BACKGROUND: Heme oxygenase-1 (HO-1) exhibits cytoprotective effects in many different cell types and is induced by nicotine exposure in human gingival fibroblasts. Nicotine 115-123 heme oxygenase 1 Homo sapiens 30-34 18205746-2 2008 However, the role of HO-1 in cancer cells exposed to nicotine has not previously been described. Nicotine 53-61 heme oxygenase 1 Homo sapiens 21-25 18205746-5 2008 RESULTS: Nicotine-induced HO-1 production and had cytotoxic effects on cells in both a concentration- and time-dependent manner. Nicotine 9-17 heme oxygenase 1 Homo sapiens 26-30 18205746-6 2008 Nicotine-induced cytotoxicity and accumulation of HO-1 were greater in IHOK cells than in HN12 cells. Nicotine 0-8 heme oxygenase 1 Homo sapiens 50-54 18205746-7 2008 Molecular inhibitors of the ERK, p38 MAP kinase, PI3 K, and NF-kappaB signaling pathways blocked the cytotoxic effects and induction of HO-1 expression by nicotine. Nicotine 155-163 heme oxygenase 1 Homo sapiens 136-140 18205746-8 2008 Treatment with antioxidants (bilirubin, N-acetylcysteine) protected cells against nicotine-induced cytotoxicity and blocked the upregulation of HO-1, the effects of which were more pronounced in IHOK cells than in HN12 cells. Bilirubin 29-38 heme oxygenase 1 Homo sapiens 144-148 18205746-8 2008 Treatment with antioxidants (bilirubin, N-acetylcysteine) protected cells against nicotine-induced cytotoxicity and blocked the upregulation of HO-1, the effects of which were more pronounced in IHOK cells than in HN12 cells. Acetylcysteine 40-56 heme oxygenase 1 Homo sapiens 144-148 18205746-9 2008 CONCLUSIONS: Collectively, these results suggest that HO-1 plays a principal role in the protective response to nicotine in oral cancer and immortalized keratinocytes. Nicotine 112-120 heme oxygenase 1 Homo sapiens 54-58 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. ferric protoporphyrin ix 37-61 heme oxygenase 1 Homo sapiens 13-17 18344023-8 2008 Both cobalt-protoporphyrin (CoPP) and CoCl2 markedly induced HO-1 expression. cobaltiprotoporphyrin 28-32 heme oxygenase 1 Homo sapiens 61-65 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. ferric protoporphyrin ix 37-61 heme oxygenase 1 Homo sapiens 220-224 18344023-8 2008 Both cobalt-protoporphyrin (CoPP) and CoCl2 markedly induced HO-1 expression. cobaltous chloride 38-43 heme oxygenase 1 Homo sapiens 61-65 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. Tetradecanoylphorbol Acetate 72-75 heme oxygenase 1 Homo sapiens 13-17 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. Tetradecanoylphorbol Acetate 72-75 heme oxygenase 1 Homo sapiens 220-224 18414232-0 2008 Hemin arginate-induced heme oxygenase 1 expression improves liver microcirculation and mediates an anti-inflammatory cytokine response after hemorrhagic shock. L-argininate 6-14 heme oxygenase 1 Homo sapiens 23-39 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. protoporphyrin IX 44-61 heme oxygenase 1 Homo sapiens 13-17 18483307-3 2008 Elevation of HO-1 gene expression by ferric protoporphyrin IX inhibited TPA-induced invasion of MCF-7 cells, which was blocked by adding the heme oxygenase inhibitor, tin protoporphyrin IX, or transfection of cells with HO-1 short hairpin RNA. protoporphyrin IX 44-61 heme oxygenase 1 Homo sapiens 220-224 18483307-4 2008 MCF-7 cells overexpressing HO-1 (MCF-7/HO-1) were established in the present study, and TPA-induced MMP-9 gene expression, tumor invasion, and colony formation were significantly reduced in MCF-7/HO-1 cells, compared with those in Neo-transfected cells. Tetradecanoylphorbol Acetate 88-91 heme oxygenase 1 Homo sapiens 27-31 18483307-4 2008 MCF-7 cells overexpressing HO-1 (MCF-7/HO-1) were established in the present study, and TPA-induced MMP-9 gene expression, tumor invasion, and colony formation were significantly reduced in MCF-7/HO-1 cells, compared with those in Neo-transfected cells. Tetradecanoylphorbol Acetate 88-91 heme oxygenase 1 Homo sapiens 33-43 18483307-4 2008 MCF-7 cells overexpressing HO-1 (MCF-7/HO-1) were established in the present study, and TPA-induced MMP-9 gene expression, tumor invasion, and colony formation were significantly reduced in MCF-7/HO-1 cells, compared with those in Neo-transfected cells. Tetradecanoylphorbol Acetate 88-91 heme oxygenase 1 Homo sapiens 39-43 18483307-5 2008 Activation of protein kinase Calpha/extracellular signal-regulated kinases/AP-1 with stimulation of reactive oxygen species production was involved in TPA-induced invasion of MCF-7 cells, which was attenuated by HO-1 protein induced by ferric protoporphyrin IX or transfection of HO-1 expression vectors. Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 212-216 18483307-5 2008 Activation of protein kinase Calpha/extracellular signal-regulated kinases/AP-1 with stimulation of reactive oxygen species production was involved in TPA-induced invasion of MCF-7 cells, which was attenuated by HO-1 protein induced by ferric protoporphyrin IX or transfection of HO-1 expression vectors. Reactive Oxygen Species 100-123 heme oxygenase 1 Homo sapiens 280-284 18483307-5 2008 Activation of protein kinase Calpha/extracellular signal-regulated kinases/AP-1 with stimulation of reactive oxygen species production was involved in TPA-induced invasion of MCF-7 cells, which was attenuated by HO-1 protein induced by ferric protoporphyrin IX or transfection of HO-1 expression vectors. Tetradecanoylphorbol Acetate 151-154 heme oxygenase 1 Homo sapiens 212-216 18483307-5 2008 Activation of protein kinase Calpha/extracellular signal-regulated kinases/AP-1 with stimulation of reactive oxygen species production was involved in TPA-induced invasion of MCF-7 cells, which was attenuated by HO-1 protein induced by ferric protoporphyrin IX or transfection of HO-1 expression vectors. Tetradecanoylphorbol Acetate 151-154 heme oxygenase 1 Homo sapiens 280-284 18483307-5 2008 Activation of protein kinase Calpha/extracellular signal-regulated kinases/AP-1 with stimulation of reactive oxygen species production was involved in TPA-induced invasion of MCF-7 cells, which was attenuated by HO-1 protein induced by ferric protoporphyrin IX or transfection of HO-1 expression vectors. ferric protoporphyrin ix 236-260 heme oxygenase 1 Homo sapiens 212-216 18414232-3 2008 Hemin arginate (HAR) induces HO-1 and protects against shock-induced organ failure. hemin arginate 0-14 heme oxygenase 1 Homo sapiens 29-33 18237271-6 2008 The consequence of this finding is that signalling initiated by electrophilic lipids differs from agonists that do not form covalent adducts with proteins because the constant generation of very lowconcentrations of 15d-PGJ2 can lead to induction of GSH or HO-1. 15-deoxy-delta(12,14)-prostaglandin J2 216-224 heme oxygenase 1 Homo sapiens 257-261 18275848-0 2008 Heme oxygenase-1 is a novel target and antioxidant mediator of S-adenosylmethionine. S-Adenosylmethionine 63-83 heme oxygenase 1 Homo sapiens 0-16 18275848-7 2008 HO-1 upregulation by SAM was causally related to a decrease in NADPH-mediated production of oxygen radicals. NADP 63-68 heme oxygenase 1 Homo sapiens 0-4 18275848-7 2008 HO-1 upregulation by SAM was causally related to a decrease in NADPH-mediated production of oxygen radicals. Reactive Oxygen Species 92-107 heme oxygenase 1 Homo sapiens 0-4 18226606-3 2008 Incubation with arsenite concentration- and time-dependently increased the expression of stress proteins, heat shock protein 70, and heme oxygenase-1 in DRG explants. arsenite 16-24 heme oxygenase 1 Homo sapiens 133-149 18048804-0 2008 Acrolein induces heme oxygenase-1 through PKC-delta and PI3K in human bronchial epithelial cells. Acrolein 0-8 heme oxygenase 1 Homo sapiens 17-33 18237271-3 2008 Protective pathways activated by 15d-PGJ2 include those controlling the synthesis of the intracellular antioxidants GSH and the enzyme HO-1 (haem oxygenase-1). 15-deoxy-delta(12,14)-prostaglandin J2 33-41 heme oxygenase 1 Homo sapiens 135-157 18390757-0 2008 Heme oxygenase-1 is a critical regulator of nitric oxide production in enterohemorrhagic Escherichia coli-infected human enterocytes. Nitric Oxide 44-56 heme oxygenase 1 Homo sapiens 0-16 18390757-8 2008 Knock-down of hmox-1 gene expression by small interfering RNA or the blockade of HO-1 activity by zinc protoporphyrin IX abrogated the EHEC-dependent inhibition of STAT-1 activation and iNOS mRNA expression in activated human enterocytes. zinc protoporphyrin 98-120 heme oxygenase 1 Homo sapiens 81-85 18048804-6 2008 Acrolein-mediated HO-1 induction was significantly attenuated by pan-protein kinase C (PKC) inhibitors RO318220, staurosporine, and PKC-delta selective inhibitor rottlerin and PKC-delta small interfering RNA. Staurosporine 113-126 heme oxygenase 1 Homo sapiens 18-22 18048804-1 2008 Heme oxygenase-1 (HO-1) catalyzes the rate limiting reaction of heme metabolism and plays critical roles in resistance to oxidative stress and other cellular functions. Heme 64-68 heme oxygenase 1 Homo sapiens 0-16 18048804-6 2008 Acrolein-mediated HO-1 induction was significantly attenuated by pan-protein kinase C (PKC) inhibitors RO318220, staurosporine, and PKC-delta selective inhibitor rottlerin and PKC-delta small interfering RNA. rottlerin 162-171 heme oxygenase 1 Homo sapiens 18-22 18048804-7 2008 The HO-1 induction was also decreased by phosphatidylinositol 3-kinase (PI3K) inhibitors LY294002 and wortmannin. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 89-97 heme oxygenase 1 Homo sapiens 4-8 18048804-1 2008 Heme oxygenase-1 (HO-1) catalyzes the rate limiting reaction of heme metabolism and plays critical roles in resistance to oxidative stress and other cellular functions. Heme 64-68 heme oxygenase 1 Homo sapiens 18-22 18048804-7 2008 The HO-1 induction was also decreased by phosphatidylinositol 3-kinase (PI3K) inhibitors LY294002 and wortmannin. Wortmannin 102-112 heme oxygenase 1 Homo sapiens 4-8 18048804-4 2008 In this investigation we studied HO-1 induction in response to acrolein and determined the signaling pathways involved in human bronchial epithelial cells (HBE1 cells). Acrolein 63-71 heme oxygenase 1 Homo sapiens 33-37 18048804-9 2008 Furthermore, Nrf2 silencing significantly attenuated the HO-1 induction by acrolein. Acrolein 75-83 heme oxygenase 1 Homo sapiens 57-61 18048804-6 2008 Acrolein-mediated HO-1 induction was significantly attenuated by pan-protein kinase C (PKC) inhibitors RO318220, staurosporine, and PKC-delta selective inhibitor rottlerin and PKC-delta small interfering RNA. Acrolein 0-8 heme oxygenase 1 Homo sapiens 18-22 18048804-11 2008 Taken together, our results indicate that acrolein up-regulates HO-1 expression through both PKC-delta and PI3K pathways in HBE1 cells; PKC-delta appears to regulate HO-1 induction via modulating Nrf2 nuclear translocation, while PI3K may work through targeting on downstream signaling molecules other than Nrf2. Acrolein 42-50 heme oxygenase 1 Homo sapiens 64-68 18048804-6 2008 Acrolein-mediated HO-1 induction was significantly attenuated by pan-protein kinase C (PKC) inhibitors RO318220, staurosporine, and PKC-delta selective inhibitor rottlerin and PKC-delta small interfering RNA. Ro 31-8220 103-111 heme oxygenase 1 Homo sapiens 18-22 18082923-3 2008 (-)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol, has been shown to induce expression of glutathione S-transferase, glutathione peroxidase, glutamate cysteine ligase, hemeoxygenase-1, etc. epigallocatechin gallate 0-30 heme oxygenase 1 Homo sapiens 186-201 18371544-3 2008 HO-1, an integral component of an important cytoprotective mechanism, mediates its action through removal of heme, the generation of heme breakdown reaction products (biliverdin, free iron, and carbon monoxide), and modulation of key cellular molecules. Heme 109-113 heme oxygenase 1 Homo sapiens 0-4 18371544-3 2008 HO-1, an integral component of an important cytoprotective mechanism, mediates its action through removal of heme, the generation of heme breakdown reaction products (biliverdin, free iron, and carbon monoxide), and modulation of key cellular molecules. Biliverdine 167-177 heme oxygenase 1 Homo sapiens 0-4 18371544-3 2008 HO-1, an integral component of an important cytoprotective mechanism, mediates its action through removal of heme, the generation of heme breakdown reaction products (biliverdin, free iron, and carbon monoxide), and modulation of key cellular molecules. Iron 184-188 heme oxygenase 1 Homo sapiens 0-4 18371544-3 2008 HO-1, an integral component of an important cytoprotective mechanism, mediates its action through removal of heme, the generation of heme breakdown reaction products (biliverdin, free iron, and carbon monoxide), and modulation of key cellular molecules. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 0-4 18371544-5 2008 The discovery of a functional guanosine thymine tandem repeat polymorphism in the promoter region of the human HO-1 gene has stimulated clinical investigations in a variety of diseases. guanosine thymine 30-47 heme oxygenase 1 Homo sapiens 111-115 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 86-103 heme oxygenase 1 Homo sapiens 138-142 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 86-103 heme oxygenase 1 Homo sapiens 138-142 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 86-96 heme oxygenase 1 Homo sapiens 28-32 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 86-96 heme oxygenase 1 Homo sapiens 138-142 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 86-96 heme oxygenase 1 Homo sapiens 138-142 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 186-196 heme oxygenase 1 Homo sapiens 28-32 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 186-196 heme oxygenase 1 Homo sapiens 138-142 18206660-9 2008 The results showed that (1) HO-1 is induced in chronic cholestatic liver disease, (2) superoxide anions time- and dose-dependently induce HO-1 activity, (3) HO-1 overexpression inhibits superoxide-anions-induced apoptosis, and (4) CO blocks superoxide-anions-induced JNK phosphorylation and caspase-9, -6, -3 activation and abolishes apoptosis but does not increase necrosis. Superoxides 186-196 heme oxygenase 1 Homo sapiens 138-142 18206660-10 2008 We conclude that HO-1 and CO protect primary hepatocytes against superoxide-anions-induced apoptosis partially via inhibition of JNK activity. Superoxides 65-75 heme oxygenase 1 Homo sapiens 17-21 18404535-0 2008 Protection of chlorophyllin against oxidative damage by inducing HO-1 and NQO1 expression mediated by PI3K/Akt and Nrf2. chlorophyllin 14-27 heme oxygenase 1 Homo sapiens 65-69 18082923-3 2008 (-)-Epigallocatechin-3-gallate (EGCG), a major green tea polyphenol, has been shown to induce expression of glutathione S-transferase, glutathione peroxidase, glutamate cysteine ligase, hemeoxygenase-1, etc. epigallocatechin gallate 32-36 heme oxygenase 1 Homo sapiens 186-201 18006113-0 2008 Suppression of Nrf2-driven heme oxygenase-1 enhances the chemosensitivity of lung cancer A549 cells toward cisplatin. Cisplatin 107-116 heme oxygenase 1 Homo sapiens 27-43 18286266-4 2008 Induction of HO-1 may confer protection by controlling intracellular levels of toxic heme, or by anti-inflammatory, anti-apoptotic, and blood flow-maintaining effects of its by-products biliverdin and CO. Heme 85-89 heme oxygenase 1 Homo sapiens 13-17 18286266-4 2008 Induction of HO-1 may confer protection by controlling intracellular levels of toxic heme, or by anti-inflammatory, anti-apoptotic, and blood flow-maintaining effects of its by-products biliverdin and CO. Biliverdine 186-196 heme oxygenase 1 Homo sapiens 13-17 18360685-8 2008 A minimal HO-1-inducing formulation was prepared and showed significant cytoprotection against H2O2-induced oxidative stress. Hydrogen Peroxide 95-99 heme oxygenase 1 Homo sapiens 10-14 18006113-5 2008 A549 cells are less susceptible to cisplatin cytotoxicity than other lung cancer cell lines, concomitant with increases in HO-1 expression and MAPK phosphorylation in a time-dependent fashion. Cisplatin 35-44 heme oxygenase 1 Homo sapiens 123-127 18006113-6 2008 Furthermore, inhibition of HO-1 by siRNA and a specific HO-1 inhibitor ZnPP augments cisplatin cytotoxicity toward A549 cells. zinc protoporphyrin 71-75 heme oxygenase 1 Homo sapiens 27-31 18006113-6 2008 Furthermore, inhibition of HO-1 by siRNA and a specific HO-1 inhibitor ZnPP augments cisplatin cytotoxicity toward A549 cells. zinc protoporphyrin 71-75 heme oxygenase 1 Homo sapiens 56-60 18006113-6 2008 Furthermore, inhibition of HO-1 by siRNA and a specific HO-1 inhibitor ZnPP augments cisplatin cytotoxicity toward A549 cells. Cisplatin 85-94 heme oxygenase 1 Homo sapiens 27-31 18006113-6 2008 Furthermore, inhibition of HO-1 by siRNA and a specific HO-1 inhibitor ZnPP augments cisplatin cytotoxicity toward A549 cells. Cisplatin 85-94 heme oxygenase 1 Homo sapiens 56-60 18006113-7 2008 Pharmacologic suppression of HO-1 activity resulted in a marked increase in the ROS generation in cisplatin-treated cells. ros 80-83 heme oxygenase 1 Homo sapiens 29-33 18006113-7 2008 Pharmacologic suppression of HO-1 activity resulted in a marked increase in the ROS generation in cisplatin-treated cells. Cisplatin 98-107 heme oxygenase 1 Homo sapiens 29-33 18006113-8 2008 In addition, pharmacologic inhibitors of MAPK suppressed the induction of HO-1 and Nrf2 expression by cisplatin. Cisplatin 102-111 heme oxygenase 1 Homo sapiens 74-78 18006113-9 2008 These findings suggest that HO-1 may modulate the chemosensitivity of lung cancer A549 cells to cisplatin through the MAPK-Nrf2 pathway. Cisplatin 96-105 heme oxygenase 1 Homo sapiens 28-32 18178725-0 2008 Heme oxygenase-1 induction depletes heme and attenuates pulmonary artery relaxation and guanylate cyclase activation by nitric oxide. Heme 36-40 heme oxygenase 1 Homo sapiens 0-16 17657593-0 2008 Structural influence of isothiocyanates on the antioxidant response element (ARE)-mediated heme oxygenase-1 (HO-1) expression. Isothiocyanates 24-39 heme oxygenase 1 Homo sapiens 91-107 17657593-0 2008 Structural influence of isothiocyanates on the antioxidant response element (ARE)-mediated heme oxygenase-1 (HO-1) expression. Isothiocyanates 24-39 heme oxygenase 1 Homo sapiens 109-113 17657593-3 2008 In this study, the effects of ten structurally related isothiocyanates on the antioxidant response element (ARE)-mediated antioxidant enzyme heme oxygenase-1 (HO-1) induction in human hepatoma HepG2-C8 cells were evaluated. Isothiocyanates 55-70 heme oxygenase 1 Homo sapiens 141-157 17657593-3 2008 In this study, the effects of ten structurally related isothiocyanates on the antioxidant response element (ARE)-mediated antioxidant enzyme heme oxygenase-1 (HO-1) induction in human hepatoma HepG2-C8 cells were evaluated. Isothiocyanates 55-70 heme oxygenase 1 Homo sapiens 159-163 17467819-5 2008 Metoprolol increased the expression of interleukin-1alpha, cyclooxygenase-1, TNF-alpha-induced protein 3, heme oxygenase 1 and granulocyte/macrophage-colony-stimulating factor. Metoprolol 0-10 heme oxygenase 1 Homo sapiens 106-175 18174021-0 2008 Galantamine and carbon monoxide protect brain microvascular endothelial cells by heme oxygenase-1 induction. Galantamine 0-11 heme oxygenase 1 Homo sapiens 81-97 18174021-0 2008 Galantamine and carbon monoxide protect brain microvascular endothelial cells by heme oxygenase-1 induction. Carbon Monoxide 16-31 heme oxygenase 1 Homo sapiens 81-97 18174021-3 2008 We hypothesized that the protective effects of galantamine would involve induction of the protective gene, heme oxygenase-1 (HO-1), in addition to enhancement of the cholinergic system. Galantamine 47-58 heme oxygenase 1 Homo sapiens 107-123 18174021-3 2008 We hypothesized that the protective effects of galantamine would involve induction of the protective gene, heme oxygenase-1 (HO-1), in addition to enhancement of the cholinergic system. Galantamine 47-58 heme oxygenase 1 Homo sapiens 125-129 18174021-5 2008 Galantamine significantly reduced H(2)O(2)-induced cell death of mvECs in association with HO-1 induction. Galantamine 0-11 heme oxygenase 1 Homo sapiens 91-95 18174021-8 2008 These data suggest that the protective effects of galantamine require NF-kappaB activation and iNOS expression, in addition to HO-1. Galantamine 50-61 heme oxygenase 1 Homo sapiens 127-131 18174021-9 2008 Likewise, carbon monoxide (CO), one of the byproducts of HO, up-regulated HO-1 and protected mvECs from oxidative stress in a similar manner. Carbon Monoxide 10-25 heme oxygenase 1 Homo sapiens 74-78 18174021-9 2008 Likewise, carbon monoxide (CO), one of the byproducts of HO, up-regulated HO-1 and protected mvECs from oxidative stress in a similar manner. Carbon Monoxide 27-29 heme oxygenase 1 Homo sapiens 74-78 18174021-10 2008 Our data demonstrate that galantamine mediates cytoprotective effects on mvECs through induction HO-1. Galantamine 26-37 heme oxygenase 1 Homo sapiens 97-101 18174022-0 2008 Prostaglandin D2 induces heme oxygenase-1 in human retinal pigment epithelial cells. Prostaglandin D2 0-16 heme oxygenase 1 Homo sapiens 25-41 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 41-45 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 41-45 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Heme 75-79 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Heme 75-79 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 103-107 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 103-107 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Carbon Monoxide 109-124 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Biliverdine 129-139 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Biliverdine 129-139 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Bilirubin 140-149 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Bilirubin 140-149 heme oxygenase 1 Homo sapiens 18-22 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 103-107 heme oxygenase 1 Homo sapiens 0-16 18174022-4 2008 Heme oxygenase-1 (HO-1) is important for iron homeostasis via catalysis of heme degradation to release iron, carbon monoxide and biliverdin/bilirubin, and may influence iron supply to the intra-erythrocyte falciparum parasites. Iron 103-107 heme oxygenase 1 Homo sapiens 18-22 18174022-7 2008 Thus, PGD(2) may contribute to the maintenance of heme homeostasis in the brain by inducing HO-1 expression. Heme 50-54 heme oxygenase 1 Homo sapiens 92-96 18178725-0 2008 Heme oxygenase-1 induction depletes heme and attenuates pulmonary artery relaxation and guanylate cyclase activation by nitric oxide. Nitric Oxide 120-132 heme oxygenase 1 Homo sapiens 0-16 18178725-2 2008 A 24-h organ culture with 0.1 mM cobalt chloride (CoCl2) or 30 microM Co-protoporphyrin IX was developed as a method of increasing HO-1 expression. cobaltous chloride 33-48 heme oxygenase 1 Homo sapiens 131-135 18178725-2 2008 A 24-h organ culture with 0.1 mM cobalt chloride (CoCl2) or 30 microM Co-protoporphyrin IX was developed as a method of increasing HO-1 expression. cobaltous chloride 50-55 heme oxygenase 1 Homo sapiens 131-135 18178725-2 2008 A 24-h organ culture with 0.1 mM cobalt chloride (CoCl2) or 30 microM Co-protoporphyrin IX was developed as a method of increasing HO-1 expression. Cobalt protoporphyrin IX 70-90 heme oxygenase 1 Homo sapiens 131-135 18178725-4 2008 Induction of HO-1 was associated with an attenuation of pulmonary arterial relaxation to the NO-donor spermine-NONOate. spermine nitric oxide complex 102-118 heme oxygenase 1 Homo sapiens 13-17 18178725-5 2008 The presence of a HO-1 inhibitor 30 microM chromium mesoporphyrin during the 24-h organ culture (but not acute treatment with this agent) reversed the attenuation of relaxation to NO seen in arteries co-cultured with agents that increased HO-1. chromium mesoporphyrin 43-65 heme oxygenase 1 Homo sapiens 18-22 18178725-5 2008 The presence of a HO-1 inhibitor 30 microM chromium mesoporphyrin during the 24-h organ culture (but not acute treatment with this agent) reversed the attenuation of relaxation to NO seen in arteries co-cultured with agents that increased HO-1. chromium mesoporphyrin 43-65 heme oxygenase 1 Homo sapiens 239-243 18178725-8 2008 However, the increase in activity seen in the presence of 1 microM spermine-NONOate was attenuated in homogenates obtained from arteries with increased HO-1. spermine nitric oxide complex 67-83 heme oxygenase 1 Homo sapiens 152-156 18178725-9 2008 Since arteries with increased HO-1 had decreased levels of superoxide detected by the chemiluminescence of 5 microM lucigenin, superoxide did not appear to be mediating the attenuation of relaxation to NO. Superoxides 59-69 heme oxygenase 1 Homo sapiens 30-34 18178725-10 2008 These data suggest that increasing HO-1 activity depletes heme, and this is associated with an attenuation of pulmonary artery relaxation and sGC activation responses to NO. Heme 58-62 heme oxygenase 1 Homo sapiens 35-39 18293301-5 2008 Under subtoxic conditions, LOOH induced intracellular hydrogen peroxide production, a decrease of glutathione content, elevated expression of the AP-1 components c-fos and c-jun as well as of the anti-apoptotic enzyme heme oxygenase 1 (HO-1). Lipid Peroxides 27-31 heme oxygenase 1 Homo sapiens 218-234 18325350-0 2008 Zinc mesoporphyrin induces rapid and marked degradation of the transcription factor Bach1 and up-regulates HO-1. zinc mesoporphyrin 0-18 heme oxygenase 1 Homo sapiens 107-111 18325350-1 2008 Heme oxygenase 1 (HO-1) is the first and rate-controlling enzyme in heme degradation. Heme 68-72 heme oxygenase 1 Homo sapiens 0-16 18325350-1 2008 Heme oxygenase 1 (HO-1) is the first and rate-controlling enzyme in heme degradation. Heme 68-72 heme oxygenase 1 Homo sapiens 18-22 18325350-3 2008 To understand how zinc mesoporphyrin (ZnMP) induces the expression of HO-1, we investigated the effects of ZnMP on Bach1 mRNA and protein levels in human hepatoma Huh-7 cells by quantitative RT-PCR and Western blots. zinc mesoporphyrin 18-36 heme oxygenase 1 Homo sapiens 70-74 18325350-3 2008 To understand how zinc mesoporphyrin (ZnMP) induces the expression of HO-1, we investigated the effects of ZnMP on Bach1 mRNA and protein levels in human hepatoma Huh-7 cells by quantitative RT-PCR and Western blots. zinc mesoporphyrin 38-42 heme oxygenase 1 Homo sapiens 70-74 18310925-4 2008 Investigation of structure-related HO-1 inducing activity suggested that the hydroxyl group at the C-23 position in the ursane skeleton is important for this activity. ursane 120-126 heme oxygenase 1 Homo sapiens 35-39 18216035-10 2008 CONCLUSIONS: The haemoglobin-HO-1 system may be required to ensure adequate regulation of the bioavailability of haeme, iron and oxygen in human endometrium. haeme 113-118 heme oxygenase 1 Homo sapiens 29-33 18216035-10 2008 CONCLUSIONS: The haemoglobin-HO-1 system may be required to ensure adequate regulation of the bioavailability of haeme, iron and oxygen in human endometrium. Iron 120-124 heme oxygenase 1 Homo sapiens 29-33 18216035-10 2008 CONCLUSIONS: The haemoglobin-HO-1 system may be required to ensure adequate regulation of the bioavailability of haeme, iron and oxygen in human endometrium. Oxygen 129-135 heme oxygenase 1 Homo sapiens 29-33 18200630-5 2008 Up-regulation of HO-1 by cobalt protoporphyrin IX significantly reduced glycosaminoglycan degradation elicited by IL-1beta in OA cartilage explants but increased glycosaminoglycan synthesis and the expression of collagen II in OA chondrocytes in primary culture, as determined by radiometric procedures, immunoblotting and immunocytochemistry. cobaltiprotoporphyrin 25-49 heme oxygenase 1 Homo sapiens 17-21 18200630-5 2008 Up-regulation of HO-1 by cobalt protoporphyrin IX significantly reduced glycosaminoglycan degradation elicited by IL-1beta in OA cartilage explants but increased glycosaminoglycan synthesis and the expression of collagen II in OA chondrocytes in primary culture, as determined by radiometric procedures, immunoblotting and immunocytochemistry. Glycosaminoglycans 72-89 heme oxygenase 1 Homo sapiens 17-21 18200630-5 2008 Up-regulation of HO-1 by cobalt protoporphyrin IX significantly reduced glycosaminoglycan degradation elicited by IL-1beta in OA cartilage explants but increased glycosaminoglycan synthesis and the expression of collagen II in OA chondrocytes in primary culture, as determined by radiometric procedures, immunoblotting and immunocytochemistry. Glycosaminoglycans 162-179 heme oxygenase 1 Homo sapiens 17-21 18325350-4 2008 We found that ZnMP markedly up-regulated HO-1 mRNA and protein levels, and rapidly and significantly decreased Bach1 protein levels by increasing degradation of Bach1 protein [half life (t(1/2)) from 19 h to 45 min], whereas ZnMP did not influence Bach1 mRNA levels. zinc mesoporphyrin 14-18 heme oxygenase 1 Homo sapiens 41-45 18325350-7 2008 In conclusion, ZnMP produces profound post-transcriptional down-regulation of Bach1 protein levels and transcriptional up-regulation of HO-1. zinc mesoporphyrin 15-19 heme oxygenase 1 Homo sapiens 136-140 18325350-8 2008 Our results indicate that ZnMP up-regulates HO-1 gene expression by markedly increasing Bach1 protein degradation in a proteasome-dependent manner. zinc mesoporphyrin 26-30 heme oxygenase 1 Homo sapiens 44-48 18293301-5 2008 Under subtoxic conditions, LOOH induced intracellular hydrogen peroxide production, a decrease of glutathione content, elevated expression of the AP-1 components c-fos and c-jun as well as of the anti-apoptotic enzyme heme oxygenase 1 (HO-1). Lipid Peroxides 27-31 heme oxygenase 1 Homo sapiens 236-240 18293301-8 2008 Application of SnPPIX, a HO-1 inhibitor, decreased the viability of HCC-1.2 cells, indicating the protective role of HO-1 induction. tin protoporphyrin IX 15-21 heme oxygenase 1 Homo sapiens 25-29 18293301-8 2008 Application of SnPPIX, a HO-1 inhibitor, decreased the viability of HCC-1.2 cells, indicating the protective role of HO-1 induction. tin protoporphyrin IX 15-21 heme oxygenase 1 Homo sapiens 117-121 18024796-0 2008 Targeting of heat shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in leukemic cells in chronic myeloid leukemia: a novel approach to overcome resistance against imatinib. Imatinib Mesylate 162-170 heme oxygenase 1 Homo sapiens 13-34 18071189-5 2008 Ox-PAPE-N-biotin, like Ox-PAPC, induced interleukin-8 (IL-8) protein synthesis and stimulated IL-8, low density lipoprotein receptor, heme oxygenase-1, and activating transcription factor-3 mRNA expression in HAECs. ox-pape 0-7 heme oxygenase 1 Homo sapiens 134-189 18071189-5 2008 Ox-PAPE-N-biotin, like Ox-PAPC, induced interleukin-8 (IL-8) protein synthesis and stimulated IL-8, low density lipoprotein receptor, heme oxygenase-1, and activating transcription factor-3 mRNA expression in HAECs. n-biotin 8-16 heme oxygenase 1 Homo sapiens 134-189 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. zinc protoporphyrine 51-71 heme oxygenase 1 Homo sapiens 20-25 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. zinc protoporphyrine 51-71 heme oxygenase 1 Homo sapiens 26-30 18024796-0 2008 Targeting of heat shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in leukemic cells in chronic myeloid leukemia: a novel approach to overcome resistance against imatinib. Imatinib Mesylate 162-170 heme oxygenase 1 Homo sapiens 36-41 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. peg-znpp 73-81 heme oxygenase 1 Homo sapiens 20-25 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. peg-znpp 73-81 heme oxygenase 1 Homo sapiens 26-30 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. 2-Butenedioic acid (2Z)-, polymer with ethenylbenzene 86-105 heme oxygenase 1 Homo sapiens 20-25 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. 2-Butenedioic acid (2Z)-, polymer with ethenylbenzene 86-105 heme oxygenase 1 Homo sapiens 26-30 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. sma-znpp 133-141 heme oxygenase 1 Homo sapiens 20-25 18024796-4 2008 Moreover, targeting Hsp32/HO-1 by either pegylated zinc protoporphyrine (PEG-ZnPP) or styrene maleic acid-micelle-encapsulated ZnPP (SMA-ZnPP) resulted in growth inhibition of BCR/ABL-transformed cells. sma-znpp 133-141 heme oxygenase 1 Homo sapiens 26-30 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. Imatinib Mesylate 76-84 heme oxygenase 1 Homo sapiens 9-14 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. Imatinib Mesylate 76-84 heme oxygenase 1 Homo sapiens 15-19 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. nilotinib 88-97 heme oxygenase 1 Homo sapiens 9-14 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. nilotinib 88-97 heme oxygenase 1 Homo sapiens 15-19 18062931-5 2008 We have previously shown that nitric oxide and S-nitrosothiols cause nuclear accumulation of Nrf2 and upregulation of the ARE-regulated gene HO-1. Nitric Oxide 30-42 heme oxygenase 1 Homo sapiens 141-145 18062931-5 2008 We have previously shown that nitric oxide and S-nitrosothiols cause nuclear accumulation of Nrf2 and upregulation of the ARE-regulated gene HO-1. S-Nitrosothiols 47-62 heme oxygenase 1 Homo sapiens 141-145 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. Imatinib Mesylate 132-140 heme oxygenase 1 Homo sapiens 9-14 18024796-0 2008 Targeting of heat shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in leukemic cells in chronic myeloid leukemia: a novel approach to overcome resistance against imatinib. Imatinib Mesylate 162-170 heme oxygenase 1 Homo sapiens 43-59 18024796-7 2008 Finally, Hsp32/HO-1-targeting compounds were found to synergize with either imatinib or nilotinib in producing growth inhibition in imatinib-resistant K562 cells and in Ba/F3 cells harboring the T315I mutant of BCR/ABL. Imatinib Mesylate 132-140 heme oxygenase 1 Homo sapiens 15-19 18024796-8 2008 In summary, these data show that HO-1 is a promising novel target in imatinib-resistant CML. Imatinib Mesylate 69-77 heme oxygenase 1 Homo sapiens 33-37 18024796-0 2008 Targeting of heat shock protein 32 (Hsp32)/heme oxygenase-1 (HO-1) in leukemic cells in chronic myeloid leukemia: a novel approach to overcome resistance against imatinib. Imatinib Mesylate 162-170 heme oxygenase 1 Homo sapiens 61-65 18215717-3 2008 In this study, we explored: 1) the HO-1 protein content and HO activity in human umbilical vein endothelial cells (HUVECs) exposed to different glucose concentrations, and 2) the mechanisms which account for the high glucose-induced effects on HO-1. Glucose 217-224 heme oxygenase 1 Homo sapiens 244-248 18215717-6 2008 At 10 mM glucose, an increase of HO-1 protein expression and HO activity was observed, whereas at 20 mM, there was no change in protein content and activity relative to at 5.5 mM glucose. Glucose 9-16 heme oxygenase 1 Homo sapiens 33-37 18215717-0 2008 Heme oxygenase-1 is an important modulator in limiting glucose-induced apoptosis in human umbilical vein endothelial cells. Glucose 55-62 heme oxygenase 1 Homo sapiens 0-16 18215717-7 2008 HO-1 protein expression in HUVECs exposed to 20 mM of glucose was increased in the presence of 20 U/ml superoxide dismutase (SOD). Glucose 54-61 heme oxygenase 1 Homo sapiens 0-4 18215717-3 2008 In this study, we explored: 1) the HO-1 protein content and HO activity in human umbilical vein endothelial cells (HUVECs) exposed to different glucose concentrations, and 2) the mechanisms which account for the high glucose-induced effects on HO-1. Glucose 144-151 heme oxygenase 1 Homo sapiens 35-39 18215717-8 2008 HO-1 gene silencing augments ROS production both at 5.5 and 10 mM glucose, leading to an increased apoptosis. Reactive Oxygen Species 29-32 heme oxygenase 1 Homo sapiens 0-4 18215717-3 2008 In this study, we explored: 1) the HO-1 protein content and HO activity in human umbilical vein endothelial cells (HUVECs) exposed to different glucose concentrations, and 2) the mechanisms which account for the high glucose-induced effects on HO-1. Glucose 217-224 heme oxygenase 1 Homo sapiens 35-39 18215717-8 2008 HO-1 gene silencing augments ROS production both at 5.5 and 10 mM glucose, leading to an increased apoptosis. Glucose 66-73 heme oxygenase 1 Homo sapiens 0-4 18215717-9 2008 We conclude that, in endothelial cells, the regulation of HO-1 by glucose is dependent upon levels of glucose itself. Glucose 66-73 heme oxygenase 1 Homo sapiens 58-62 18215717-9 2008 We conclude that, in endothelial cells, the regulation of HO-1 by glucose is dependent upon levels of glucose itself. Glucose 102-109 heme oxygenase 1 Homo sapiens 58-62 18036570-0 2008 Anti-inflammatory effects of HO-1 activity in vascular endothelial cells, commentary on "Carbon monoxide donors or heme oxygenase (HO-1) overexpression blocks interleukin-18-mediated NF-kappaB-PTEN-dependent human cardiac endothelial cell death". Carbon Monoxide 89-104 heme oxygenase 1 Homo sapiens 29-33 18021765-6 2008 While LY294002, a non-selective phosphotidylinositol 3-kinase (PI3K) inhibitor, was able to reduce brazilin-induced phosphorylation of Akt and the subsequent induction of HO-1. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 6-14 heme oxygenase 1 Homo sapiens 171-175 18021765-10 2008 These results demonstrate that the expression of HO-1 by brazilin is mediated via the PI3K/Akt and ERK pathways, and this expression inhibits t-BHP-induced cell death in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. brazilin 57-65 heme oxygenase 1 Homo sapiens 49-53 18021765-10 2008 These results demonstrate that the expression of HO-1 by brazilin is mediated via the PI3K/Akt and ERK pathways, and this expression inhibits t-BHP-induced cell death in House Ear Institute-Organ of Corti 1 (HEI-OC1) cells. tert-Butylhydroperoxide 142-147 heme oxygenase 1 Homo sapiens 49-53 18070969-8 2008 One gene whose expression was affected by SbV was the heme oxygenase gene HMOX-1, and this change was observed both in the monocytic cell line THP-1 and in primary human monocyte-derived macrophages. SBV 42-45 heme oxygenase 1 Homo sapiens 74-80 18215737-1 2008 The objective of this study was to determine whether heme oxygenase-1 (HO-1) or heme metabolites exert cytoprotective effects on interleukin-18-mediated endothelial cell (EC) death. Heme 53-57 heme oxygenase 1 Homo sapiens 71-75 18215737-6 2008 Furthermore, hemin induced HO-1 expression, and HO-1 knockdown, HO-1 inhibition, or CO scavengers all reversed the prosurvival effects of hemin. Hemin 13-18 heme oxygenase 1 Homo sapiens 27-31 18819247-8 2008 Dose-dependent, high-level HO-1 protein expression was achieved following culture of NPICC in medium containing either cobalt protoporphyrin (CoPP) or cobalt mesoporphyrin (CoMP). cobaltiprotoporphyrin 119-140 heme oxygenase 1 Homo sapiens 27-31 18045827-2 2008 A functional dinucleotide repeat (GT)(n) polymorphism, within the HO-1 promoter, regulates HO-1 gene expression; a short number of repeats (S-allele <25) increases transcription. Dinucleoside Phosphates 13-25 heme oxygenase 1 Homo sapiens 66-70 18045827-2 2008 A functional dinucleotide repeat (GT)(n) polymorphism, within the HO-1 promoter, regulates HO-1 gene expression; a short number of repeats (S-allele <25) increases transcription. Dinucleoside Phosphates 13-25 heme oxygenase 1 Homo sapiens 91-95 17980396-4 2008 Exposure of RAW 264.7 macrophages to HOCl resulted in increased protein levels of Nrf2 in nuclear extractions, as well as a time- and dose-dependent increase in the expression of Nrf2 target genes, including heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 (NQO-1), glutamate cysteine ligase catalytic subunit (GCLC), and glutathione synthetase (GS). Hypochlorous Acid 37-41 heme oxygenase 1 Homo sapiens 208-224 18042465-3 2008 Cobalt protoporphyrin (CoPP) induced the expression of both HO-1 and p53 without significant toxicity to the cells. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 60-64 18042465-3 2008 Cobalt protoporphyrin (CoPP) induced the expression of both HO-1 and p53 without significant toxicity to the cells. cobaltiprotoporphyrin 23-27 heme oxygenase 1 Homo sapiens 60-64 18042465-4 2008 In addition, the blockage of HO activity with the iron chelator DFO or with HO-1 siRNA inhibited the CoPP-induced expression of p53. cobaltiprotoporphyrin 101-105 heme oxygenase 1 Homo sapiens 76-80 18195184-3 2008 Recent work has identified heme oxygenase-1 and its gaseous product, carbon monoxide, to possess potent proangiogenic properties in addition to well-recognized antiinflammatory, antioxidant, and antiapoptotic effects. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 27-43 18819247-8 2008 Dose-dependent, high-level HO-1 protein expression was achieved following culture of NPICC in medium containing either cobalt protoporphyrin (CoPP) or cobalt mesoporphyrin (CoMP). cobaltiprotoporphyrin 142-146 heme oxygenase 1 Homo sapiens 27-31 18819247-8 2008 Dose-dependent, high-level HO-1 protein expression was achieved following culture of NPICC in medium containing either cobalt protoporphyrin (CoPP) or cobalt mesoporphyrin (CoMP). cobalt mesoporphyrin 151-171 heme oxygenase 1 Homo sapiens 27-31 18819247-8 2008 Dose-dependent, high-level HO-1 protein expression was achieved following culture of NPICC in medium containing either cobalt protoporphyrin (CoPP) or cobalt mesoporphyrin (CoMP). cobalt mesoporphyrin 173-177 heme oxygenase 1 Homo sapiens 27-31 18289069-3 2008 The precise underlying mechanisms for HO-1-based protection are not yet completely understood, but appear to involve the protective effects of HO-1 by-products, carbon monoxide (CO), biliverdin/bilirubin and free iron. Carbon Monoxide 161-176 heme oxygenase 1 Homo sapiens 38-42 18289069-3 2008 The precise underlying mechanisms for HO-1-based protection are not yet completely understood, but appear to involve the protective effects of HO-1 by-products, carbon monoxide (CO), biliverdin/bilirubin and free iron. Carbon Monoxide 178-180 heme oxygenase 1 Homo sapiens 38-42 18289069-3 2008 The precise underlying mechanisms for HO-1-based protection are not yet completely understood, but appear to involve the protective effects of HO-1 by-products, carbon monoxide (CO), biliverdin/bilirubin and free iron. Iron 213-217 heme oxygenase 1 Homo sapiens 38-42 18289069-3 2008 The precise underlying mechanisms for HO-1-based protection are not yet completely understood, but appear to involve the protective effects of HO-1 by-products, carbon monoxide (CO), biliverdin/bilirubin and free iron. Biliverdine 183-193 heme oxygenase 1 Homo sapiens 38-42 18289069-5 2008 There is evidence supporting that HO-1-derived CO also interacts with other gaseous molecules, such as nitric oxide (NO) and hydrogen sulfide (H2S) that may relate to either vascular protection or injury. Nitric Oxide 103-115 heme oxygenase 1 Homo sapiens 34-38 18289069-5 2008 There is evidence supporting that HO-1-derived CO also interacts with other gaseous molecules, such as nitric oxide (NO) and hydrogen sulfide (H2S) that may relate to either vascular protection or injury. Hydrogen Sulfide 125-141 heme oxygenase 1 Homo sapiens 34-38 18289069-3 2008 The precise underlying mechanisms for HO-1-based protection are not yet completely understood, but appear to involve the protective effects of HO-1 by-products, carbon monoxide (CO), biliverdin/bilirubin and free iron. Bilirubin 194-203 heme oxygenase 1 Homo sapiens 38-42 18289069-5 2008 There is evidence supporting that HO-1-derived CO also interacts with other gaseous molecules, such as nitric oxide (NO) and hydrogen sulfide (H2S) that may relate to either vascular protection or injury. Hydrogen Sulfide 143-146 heme oxygenase 1 Homo sapiens 34-38 17898018-9 2008 Interestingly, sevoflurane treatment also resulted in an enhanced expression of HSP-32. Sevoflurane 15-26 heme oxygenase 1 Homo sapiens 80-86 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 0-16 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 18-22 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 0-16 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 18-22 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Iron 107-111 heme oxygenase 1 Homo sapiens 0-16 18289070-1 2008 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to generate carbon monoxide, biliverdin and free iron. Iron 107-111 heme oxygenase 1 Homo sapiens 18-22 18289070-4 2008 However, experimental observations indicate that the extent of HO-1 induction may be critical because excessive heme degradation may result in toxic levels of CO, bilirubin and, more importantly, iron. Heme 112-116 heme oxygenase 1 Homo sapiens 63-67 18289070-4 2008 However, experimental observations indicate that the extent of HO-1 induction may be critical because excessive heme degradation may result in toxic levels of CO, bilirubin and, more importantly, iron. Carbon Monoxide 159-161 heme oxygenase 1 Homo sapiens 63-67 18289070-4 2008 However, experimental observations indicate that the extent of HO-1 induction may be critical because excessive heme degradation may result in toxic levels of CO, bilirubin and, more importantly, iron. Bilirubin 163-172 heme oxygenase 1 Homo sapiens 63-67 18289070-4 2008 However, experimental observations indicate that the extent of HO-1 induction may be critical because excessive heme degradation may result in toxic levels of CO, bilirubin and, more importantly, iron. Iron 196-200 heme oxygenase 1 Homo sapiens 63-67 18289072-1 2008 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme of ferroheme metabolic pathway, which has the functions of anti-oxidation, anti-inflammatory, anti-apoptosis and anti-smooth muscle hyperplasia. Heme 55-64 heme oxygenase 1 Homo sapiens 0-16 18289072-1 2008 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme of ferroheme metabolic pathway, which has the functions of anti-oxidation, anti-inflammatory, anti-apoptosis and anti-smooth muscle hyperplasia. Heme 55-64 heme oxygenase 1 Homo sapiens 18-22 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Heme 83-87 heme oxygenase 1 Homo sapiens 0-16 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Heme 83-87 heme oxygenase 1 Homo sapiens 18-22 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 0-16 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 18-22 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Iron 110-114 heme oxygenase 1 Homo sapiens 0-16 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Iron 110-114 heme oxygenase 1 Homo sapiens 18-22 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 0-16 18289074-1 2008 Heme oxygenase-1 (HO-1) is an inducible rate-limiting enzyme which catalyzes group heme into carbon monoxide, iron and bilirubin. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 18-22 20020861-6 2008 Induction of the HMOX1 was detected with a dose of arsenite at as low as 0.3 muM (0.04 ppm) and reached its maximum at 4 h after the treatment. arsenite 51-59 heme oxygenase 1 Homo sapiens 17-22 18771089-4 2008 When HepG2 cells were incubated with non-toxic concentrations of delta-aminolevulinic acid, protoporphyrin IX, or pheophorbide a and then exposed to visible light for 90 min, the mRNA level of HO-1 began increasing markedly, reaching the maximal level in 4 h. Following the transient induction of HO-1, the mRNA level of ABCG2 gradually increased in a time-dependent manner, whereas the ABCB6 mRNA level was little affected. Aminolevulinic Acid 65-90 heme oxygenase 1 Homo sapiens 193-197 18771089-4 2008 When HepG2 cells were incubated with non-toxic concentrations of delta-aminolevulinic acid, protoporphyrin IX, or pheophorbide a and then exposed to visible light for 90 min, the mRNA level of HO-1 began increasing markedly, reaching the maximal level in 4 h. Following the transient induction of HO-1, the mRNA level of ABCG2 gradually increased in a time-dependent manner, whereas the ABCB6 mRNA level was little affected. protoporphyrin IX 92-109 heme oxygenase 1 Homo sapiens 193-197 18771089-4 2008 When HepG2 cells were incubated with non-toxic concentrations of delta-aminolevulinic acid, protoporphyrin IX, or pheophorbide a and then exposed to visible light for 90 min, the mRNA level of HO-1 began increasing markedly, reaching the maximal level in 4 h. Following the transient induction of HO-1, the mRNA level of ABCG2 gradually increased in a time-dependent manner, whereas the ABCB6 mRNA level was little affected. pheophorbide a 114-128 heme oxygenase 1 Homo sapiens 193-197 18771089-4 2008 When HepG2 cells were incubated with non-toxic concentrations of delta-aminolevulinic acid, protoporphyrin IX, or pheophorbide a and then exposed to visible light for 90 min, the mRNA level of HO-1 began increasing markedly, reaching the maximal level in 4 h. Following the transient induction of HO-1, the mRNA level of ABCG2 gradually increased in a time-dependent manner, whereas the ABCB6 mRNA level was little affected. pheophorbide a 114-128 heme oxygenase 1 Homo sapiens 297-301 18771089-6 2008 On the other hand, the mRNA level of HO-1 was remarkably enhanced by Zn(2+)-protoporphyrin IX or hemin even in the absence of light. zn(2+)-protoporphyrin ix 69-93 heme oxygenase 1 Homo sapiens 37-41 20020861-7 2008 The arsenite-induced HMOX1 expression was attenuated by the promoted glutathione (GSH) synthesis by N-acetyl-L-cysteine (NAC). arsenite 4-12 heme oxygenase 1 Homo sapiens 21-26 20020861-7 2008 The arsenite-induced HMOX1 expression was attenuated by the promoted glutathione (GSH) synthesis by N-acetyl-L-cysteine (NAC). Glutathione 69-80 heme oxygenase 1 Homo sapiens 21-26 20020861-7 2008 The arsenite-induced HMOX1 expression was attenuated by the promoted glutathione (GSH) synthesis by N-acetyl-L-cysteine (NAC). Glutathione 82-85 heme oxygenase 1 Homo sapiens 21-26 20020861-9 2008 Thus, induction of HMOX1 gene is highly sensitive and also selective against arsenite in the cells. arsenite 77-85 heme oxygenase 1 Homo sapiens 19-24 18165777-3 2007 METHOD: We studied the impact of HO-1 induction with cobalt protoporphyrin (CoPP) in experimental pancreas transplantation with moderate (6 hr) and prolonged (20 hr) cold ischemic time (CIT). cobaltiprotoporphyrin 53-74 heme oxygenase 1 Homo sapiens 33-37 17965015-2 2007 The crystal structures of apo- and heme-bound truncated human HO-2 reveal a primarily alpha-helical architecture similar to that of human HO-1 and other known HOs. Heme 35-39 heme oxygenase 1 Homo sapiens 138-142 18165777-3 2007 METHOD: We studied the impact of HO-1 induction with cobalt protoporphyrin (CoPP) in experimental pancreas transplantation with moderate (6 hr) and prolonged (20 hr) cold ischemic time (CIT). cobaltiprotoporphyrin 76-80 heme oxygenase 1 Homo sapiens 33-37 18165777-9 2007 CoPP pretreatment markedly increased HO-1 gene expression in donor pancreas (130-fold increase) by means of quantitative reverse transcriptase -polymerase chain reaction. cobaltiprotoporphyrin 0-4 heme oxygenase 1 Homo sapiens 37-41 18026199-10 2007 Treatment with hemin, a well-known specific inducer of HO-1, led to clear nuclear localisation of HO-1 in both cell lines and highly induced HO-1 expression in both cellular compartments. Hemin 15-20 heme oxygenase 1 Homo sapiens 55-59 18026199-10 2007 Treatment with hemin, a well-known specific inducer of HO-1, led to clear nuclear localisation of HO-1 in both cell lines and highly induced HO-1 expression in both cellular compartments. Hemin 15-20 heme oxygenase 1 Homo sapiens 98-102 18026199-10 2007 Treatment with hemin, a well-known specific inducer of HO-1, led to clear nuclear localisation of HO-1 in both cell lines and highly induced HO-1 expression in both cellular compartments. Hemin 15-20 heme oxygenase 1 Homo sapiens 98-102 17883332-2 2007 We examined, in vascular endothelial cells, whether the selective expression of heme oxygenase-1 (HO-1) offers cytoprotection against glucose- and TNF-mediated cell death. Glucose 134-141 heme oxygenase 1 Homo sapiens 80-96 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Heme 51-55 heme oxygenase 1 Homo sapiens 0-16 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Heme 51-55 heme oxygenase 1 Homo sapiens 18-22 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 89-104 heme oxygenase 1 Homo sapiens 0-16 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 89-104 heme oxygenase 1 Homo sapiens 18-22 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Iron 148-152 heme oxygenase 1 Homo sapiens 88-104 17883332-2 2007 We examined, in vascular endothelial cells, whether the selective expression of heme oxygenase-1 (HO-1) offers cytoprotection against glucose- and TNF-mediated cell death. Glucose 134-141 heme oxygenase 1 Homo sapiens 98-102 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Iron 148-152 heme oxygenase 1 Homo sapiens 106-110 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 88-104 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 106-110 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Bilirubin 196-205 heme oxygenase 1 Homo sapiens 88-104 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Bilirubin 196-205 heme oxygenase 1 Homo sapiens 106-110 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Heme 39-43 heme oxygenase 1 Homo sapiens 57-61 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 106-108 heme oxygenase 1 Homo sapiens 0-16 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Carbon Monoxide 106-108 heme oxygenase 1 Homo sapiens 18-22 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Biliverdine 111-121 heme oxygenase 1 Homo sapiens 0-16 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Biliverdine 111-121 heme oxygenase 1 Homo sapiens 18-22 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Iron 127-139 heme oxygenase 1 Homo sapiens 0-16 17822372-1 2007 Heme oxygenase-1 (HO-1) catalyzes the oxidation of heme to biologically active products: carbon monoxide (CO), biliverdin, and ferrous iron. Iron 127-139 heme oxygenase 1 Homo sapiens 18-22 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 166-168 heme oxygenase 1 Homo sapiens 57-61 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 39-55 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 57-61 17883332-6 2007 Selective expression of HO-1 prevented TNF- and hyperglycemia-mediated superoxide (O2-) formation, DNA degeneration, and upregulation of caspase, but increased the expression of pAkt and Bcl-xL, proteins responsible for endothelial dysfunction in diabetes. Superoxides 71-81 heme oxygenase 1 Homo sapiens 24-28 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Biliverdine 171-181 heme oxygenase 1 Homo sapiens 39-55 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Biliverdine 171-181 heme oxygenase 1 Homo sapiens 57-61 17883332-6 2007 Selective expression of HO-1 prevented TNF- and hyperglycemia-mediated superoxide (O2-) formation, DNA degeneration, and upregulation of caspase, but increased the expression of pAkt and Bcl-xL, proteins responsible for endothelial dysfunction in diabetes. Superoxides 83-85 heme oxygenase 1 Homo sapiens 24-28 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Iron 192-196 heme oxygenase 1 Homo sapiens 39-55 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Iron 192-196 heme oxygenase 1 Homo sapiens 57-61 17979524-6 2007 Prior exposure of cells to N-acetyl-L -cysteine blocked not only the ROS production but also the nuclear translocation of Nrf2 and its ARE binding, as well as HO-1 induction by capsaicin. Acetylcysteine 27-47 heme oxygenase 1 Homo sapiens 159-163 17979524-6 2007 Prior exposure of cells to N-acetyl-L -cysteine blocked not only the ROS production but also the nuclear translocation of Nrf2 and its ARE binding, as well as HO-1 induction by capsaicin. Capsaicin 177-186 heme oxygenase 1 Homo sapiens 159-163 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Heme 12-16 heme oxygenase 1 Homo sapiens 88-104 17979524-7 2007 Immunoblot analysis showed that whereas the level of HO-1 protein was elevated, that of NAD(P)H:quinone oxidoreductase (NQO1) was decreased after the treatment with capsaicin or the inhibitor of NQO1, dicumarol. Capsaicin 165-174 heme oxygenase 1 Homo sapiens 53-57 17979524-7 2007 Immunoblot analysis showed that whereas the level of HO-1 protein was elevated, that of NAD(P)H:quinone oxidoreductase (NQO1) was decreased after the treatment with capsaicin or the inhibitor of NQO1, dicumarol. Dicumarol 201-210 heme oxygenase 1 Homo sapiens 53-57 17919067-7 2007 Accumulating evidence indicates that biliverdin/bilirubin can mediate the protective effects of HO-1 in many disease models, such as IRI and organ transplantation, via its antiinflammatory, antiapoptotic, antiproliferative, and antioxidant properties, as well as its effects on the immune response. Biliverdine 37-47 heme oxygenase 1 Homo sapiens 96-100 17919067-7 2007 Accumulating evidence indicates that biliverdin/bilirubin can mediate the protective effects of HO-1 in many disease models, such as IRI and organ transplantation, via its antiinflammatory, antiapoptotic, antiproliferative, and antioxidant properties, as well as its effects on the immune response. Bilirubin 48-57 heme oxygenase 1 Homo sapiens 96-100 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Heme 12-16 heme oxygenase 1 Homo sapiens 106-110 17979524-0 2007 Capsaicin induces heme oxygenase-1 expression in HepG2 cells via activation of PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target. Capsaicin 0-9 heme oxygenase 1 Homo sapiens 18-34 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 88-104 17979524-0 2007 Capsaicin induces heme oxygenase-1 expression in HepG2 cells via activation of PI3K-Nrf2 signaling: NAD(P)H:quinone oxidoreductase as a potential target. nad(p)h 100-107 heme oxygenase 1 Homo sapiens 18-34 17979524-3 2007 In the present study, we found that capsaicin induced expression of heme oxygenase-1 (HO-1) in HepG2 cells. Capsaicin 36-45 heme oxygenase 1 Homo sapiens 68-84 17979524-3 2007 In the present study, we found that capsaicin induced expression of heme oxygenase-1 (HO-1) in HepG2 cells. Capsaicin 36-45 heme oxygenase 1 Homo sapiens 86-90 18077580-5 2007 Furthermore, NAC inhibited arsenite-induced elevation in the expression of stress proteins, such as heat shock protein 70 and heme oxygenase 1, as well as arsenite-induced phosphorylation of p38 mitogen-activated protein kinase. Acetylcysteine 13-16 heme oxygenase 1 Homo sapiens 126-142 18077580-5 2007 Furthermore, NAC inhibited arsenite-induced elevation in the expression of stress proteins, such as heat shock protein 70 and heme oxygenase 1, as well as arsenite-induced phosphorylation of p38 mitogen-activated protein kinase. arsenite 27-35 heme oxygenase 1 Homo sapiens 126-142 17887916-3 2007 Physiologic heme degradation is catalyzed by two functional isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, yielding carbon monoxide, iron, and biliverdin, an immediate precursor to bilirubin. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 106-110 17919067-2 2007 The inducible form of these enzymes is heme oxygenase-1 (HO-1), which is the rate-limiting enzyme that can degrade heme into equimolar quantities of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 166-168 heme oxygenase 1 Homo sapiens 39-55 17982681-0 2007 Inhibition of heme oxygenase-1 increases responsiveness of melanoma cells to ALA-based photodynamic therapy. 5-amino levulinic acid 77-80 heme oxygenase 1 Homo sapiens 14-30 17924972-9 2007 The inductive effects of SP on HO-1 and CCL20 were enhanced by HO-1 inducer hemin and the membrane-permeable guanosine 3",5"-monophosphate (cGMP) analogue 8-bromo-cGMP. Cyclic GMP 109-138 heme oxygenase 1 Homo sapiens 31-35 17924972-9 2007 The inductive effects of SP on HO-1 and CCL20 were enhanced by HO-1 inducer hemin and the membrane-permeable guanosine 3",5"-monophosphate (cGMP) analogue 8-bromo-cGMP. Cyclic GMP 140-144 heme oxygenase 1 Homo sapiens 31-35 17924972-9 2007 The inductive effects of SP on HO-1 and CCL20 were enhanced by HO-1 inducer hemin and the membrane-permeable guanosine 3",5"-monophosphate (cGMP) analogue 8-bromo-cGMP. 8-bromocyclic GMP 155-167 heme oxygenase 1 Homo sapiens 31-35 17924972-10 2007 Conversely, this pathway was inhibited by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) and the selective inhibitor of guanylate cyclase, 1H-(1,2,4)oxadiazole(4,3-a)quinoxalin-1-one (ODQ). zinc protoporphyrin 61-83 heme oxygenase 1 Homo sapiens 46-50 17924972-10 2007 Conversely, this pathway was inhibited by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) and the selective inhibitor of guanylate cyclase, 1H-(1,2,4)oxadiazole(4,3-a)quinoxalin-1-one (ODQ). zinc protoporphyrin 85-89 heme oxygenase 1 Homo sapiens 46-50 17924972-10 2007 Conversely, this pathway was inhibited by the HO-1 inhibitor zinc protoporphyrin IX (ZnPP IX) and the selective inhibitor of guanylate cyclase, 1H-(1,2,4)oxadiazole(4,3-a)quinoxalin-1-one (ODQ). 1H-(1,2,3)oxadiazolo(4,4-a)quinoxalin-1-one 189-192 heme oxygenase 1 Homo sapiens 46-50 18210237-0 2007 Costunolide inhibits production of tumor necrosis factor-alpha and interleukin-6 by inducing heme oxygenase-1 in RAW264.7 macrophages. costunolide 0-11 heme oxygenase 1 Homo sapiens 93-109 17982681-5 2007 Thus, the production and degradation of PPIX (via heme by HO-1) were simultaneously enhanced, leading to a reduced intracellular concentration of the photodynamically active substance PPIX. Heme 50-54 heme oxygenase 1 Homo sapiens 58-62 17982681-5 2007 Thus, the production and degradation of PPIX (via heme by HO-1) were simultaneously enhanced, leading to a reduced intracellular concentration of the photodynamically active substance PPIX. protoporphyrin IX 184-188 heme oxygenase 1 Homo sapiens 58-62 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. tin protoporphyrin IX 48-69 heme oxygenase 1 Homo sapiens 12-16 17973866-2 2007 Heme compounds, like hemin, a heme oxygenase-1 inducer, are used in the treatment of acute porphyria treatment. Heme 0-4 heme oxygenase 1 Homo sapiens 30-46 17973866-2 2007 Heme compounds, like hemin, a heme oxygenase-1 inducer, are used in the treatment of acute porphyria treatment. Hemin 21-26 heme oxygenase 1 Homo sapiens 30-46 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. tin protoporphyrin IX 48-69 heme oxygenase 1 Homo sapiens 33-37 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 30-51 heme oxygenase 1 Homo sapiens 84-100 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. tin protoporphyrin IX 71-77 heme oxygenase 1 Homo sapiens 12-16 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. tin protoporphyrin IX 71-77 heme oxygenase 1 Homo sapiens 33-37 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 30-51 heme oxygenase 1 Homo sapiens 102-106 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. protoporphyrin IX 73-77 heme oxygenase 1 Homo sapiens 12-16 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 53-56 heme oxygenase 1 Homo sapiens 84-100 17982681-6 2007 Diminishing HO-1 activity by the HO-1 inhibitor tin protoporphyrin IX (SnPPIX) significantly enhanced the formation of PPIX up to 1.8 fold. protoporphyrin IX 73-77 heme oxygenase 1 Homo sapiens 33-37 17982681-9 2007 The pharmacological inhibition of HO-1 activity by SnPPIX leads to a considerable increase in the sensitivity of tumor cells to ALA-PDT treatment. tin protoporphyrin IX 51-57 heme oxygenase 1 Homo sapiens 34-38 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 53-56 heme oxygenase 1 Homo sapiens 102-106 17982681-9 2007 The pharmacological inhibition of HO-1 activity by SnPPIX leads to a considerable increase in the sensitivity of tumor cells to ALA-PDT treatment. 5-amino levulinic acid 128-131 heme oxygenase 1 Homo sapiens 34-38 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 53-56 heme oxygenase 1 Homo sapiens 148-152 17982681-13 2007 The results presented indicate that HO-1 can play a protective role against ALA-PDT mediated cytotoxicity so that a specific inhibition of HO-1 activity and/or expression might be used to increase the efficacy of ALA-based photodynamic therapy. 5-amino levulinic acid 76-79 heme oxygenase 1 Homo sapiens 36-40 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 177-198 heme oxygenase 1 Homo sapiens 84-100 17982681-13 2007 The results presented indicate that HO-1 can play a protective role against ALA-PDT mediated cytotoxicity so that a specific inhibition of HO-1 activity and/or expression might be used to increase the efficacy of ALA-based photodynamic therapy. 5-amino levulinic acid 76-79 heme oxygenase 1 Homo sapiens 139-143 17982681-13 2007 The results presented indicate that HO-1 can play a protective role against ALA-PDT mediated cytotoxicity so that a specific inhibition of HO-1 activity and/or expression might be used to increase the efficacy of ALA-based photodynamic therapy. 5-amino levulinic acid 213-216 heme oxygenase 1 Homo sapiens 36-40 17982681-13 2007 The results presented indicate that HO-1 can play a protective role against ALA-PDT mediated cytotoxicity so that a specific inhibition of HO-1 activity and/or expression might be used to increase the efficacy of ALA-based photodynamic therapy. 5-amino levulinic acid 213-216 heme oxygenase 1 Homo sapiens 139-143 17982681-1 2007 Based on the observation that 5-aminolevulinic acid (ALA) induces the expression of heme oxygenase-1 (HO-1) in cultured melanoma cells, the role of HO-1 on the effectiveness of 5-aminolevulinic acid-based photodynamic therapy (ALA-PDT) was examined. 5-amino levulinic acid 177-198 heme oxygenase 1 Homo sapiens 148-152 17982681-2 2007 Transcriptional activation of the HO-1 gene is considered to be an adaptive response to oxidative and cellular stress and confers a protective capacity against cell and tissue injury, which could affect the responsiveness to ALA-PDT. 5-amino levulinic acid 225-228 heme oxygenase 1 Homo sapiens 34-38 17982681-5 2007 Thus, the production and degradation of PPIX (via heme by HO-1) were simultaneously enhanced, leading to a reduced intracellular concentration of the photodynamically active substance PPIX. protoporphyrin IX 40-44 heme oxygenase 1 Homo sapiens 58-62 17761847-1 2007 To enhance our understanding of the physiological roles of heme oxygenase (HO) isozymes, HO-1 (inducible) and HO-2 (constitutive), we developed novel imidazole-based HO inhibitors. imidazole 150-159 heme oxygenase 1 Homo sapiens 89-93 17881360-1 2007 Heme oxygenase-1 (HO-1), an inducible enzyme that metabolizes the heme group, is highly expressed in human Kaposi sarcoma lesions. Heme 66-70 heme oxygenase 1 Homo sapiens 0-16 17927807-7 2007 HO-1 inhibition, with small interfering RNA (siRNA) or zinc protoporphyrin IX, abrogated atorvastatin-mediated cytoprotection. protoporphyrin IX 60-77 heme oxygenase 1 Homo sapiens 0-4 17927807-7 2007 HO-1 inhibition, with small interfering RNA (siRNA) or zinc protoporphyrin IX, abrogated atorvastatin-mediated cytoprotection. Atorvastatin 89-101 heme oxygenase 1 Homo sapiens 0-4 17927807-9 2007 Iron chelation, adenoviral-mediated overexpression of ferritin or supplementation of culture media with biliverdin reversed the inhibitory effects of HO-1 and KLF2 siRNA, suggesting that bile pigments and ferritin mediate the antioxidant actions of statin-induced HO-1. Biliverdine 104-114 heme oxygenase 1 Homo sapiens 150-154 17927807-9 2007 Iron chelation, adenoviral-mediated overexpression of ferritin or supplementation of culture media with biliverdin reversed the inhibitory effects of HO-1 and KLF2 siRNA, suggesting that bile pigments and ferritin mediate the antioxidant actions of statin-induced HO-1. Biliverdine 104-114 heme oxygenase 1 Homo sapiens 264-268 17927807-10 2007 CONCLUSIONS: We have identified a novel link between KLF2 and HO-1 in human vascular ECs, demonstrating that atorvastatin-mediated HO-1 upregulation, and its associated antioxidant effect, is KLF2-dependent. Atorvastatin 109-121 heme oxygenase 1 Homo sapiens 62-66 17927807-10 2007 CONCLUSIONS: We have identified a novel link between KLF2 and HO-1 in human vascular ECs, demonstrating that atorvastatin-mediated HO-1 upregulation, and its associated antioxidant effect, is KLF2-dependent. Atorvastatin 109-121 heme oxygenase 1 Homo sapiens 131-135 18089404-1 2007 PURPOSE: The heme oxygenase-1 (HO-1) system is associated with the rate-limiting step of conversion of heme, one of the most critical roles in cytoprotective mechanisms. Heme 13-17 heme oxygenase 1 Homo sapiens 31-35 18089404-10 2007 After using ZnPP, an inhibitor of HO-1, the rate of Jurkat T-cell adhesion recovered to 29.08%. zinc protoporphyrin 12-16 heme oxygenase 1 Homo sapiens 34-38 17885094-0 2007 Chondroitin sulfate protects SH-SY5Y cells from oxidative stress by inducing heme oxygenase-1 via phosphatidylinositol 3-kinase/Akt. Chondroitin Sulfates 0-19 heme oxygenase 1 Homo sapiens 77-93 17885094-7 2007 CS also increased the expression of phosphorylated Akt and heme oxygenase-1 by 2-fold. Chondroitin Sulfates 0-2 heme oxygenase 1 Homo sapiens 59-75 17885094-9 2007 Taken together, these results show that CS can protect SH-SY5Y cells under oxidative stress conditions by activating protein kinase C, which phosphorylates Akt that, via the phosphatidylinositol 3-kinase/Akt pathway, induces the synthesis of the antioxidant protein heme oxygenase-1. Chondroitin Sulfates 40-42 heme oxygenase 1 Homo sapiens 266-282 17927807-4 2007 METHODS/RESULTS: Treatment of human umbilical vein and aortic ECs with atorvastatin significantly upregulated HO-1 promoter activity, mRNA expression and protein expression, increasing HO-1 enzymatic activity as shown by raised intracellular bilirubin IXalpha. Atorvastatin 71-83 heme oxygenase 1 Homo sapiens 110-114 17927807-4 2007 METHODS/RESULTS: Treatment of human umbilical vein and aortic ECs with atorvastatin significantly upregulated HO-1 promoter activity, mRNA expression and protein expression, increasing HO-1 enzymatic activity as shown by raised intracellular bilirubin IXalpha. Atorvastatin 71-83 heme oxygenase 1 Homo sapiens 185-189 17976119-8 2007 However, the anti-apoptotic action of HO-1 was reversed by SnPP. S-Nitroso-N-propionyl-D,L-penicillamine 59-63 heme oxygenase 1 Homo sapiens 38-42 17881360-1 2007 Heme oxygenase-1 (HO-1), an inducible enzyme that metabolizes the heme group, is highly expressed in human Kaposi sarcoma lesions. Heme 66-70 heme oxygenase 1 Homo sapiens 18-22 17881360-9 2007 Likewise, inhibition of HO-1 activity by chronic administration of the HO-1 inhibitor tin protoporphyrin IX to mice reduces RhoAQL-induced tumor growth by 70%. tin protoporphyrin IX 86-107 heme oxygenase 1 Homo sapiens 71-75 17881360-9 2007 Likewise, inhibition of HO-1 activity by chronic administration of the HO-1 inhibitor tin protoporphyrin IX to mice reduces RhoAQL-induced tumor growth by 70%. rhoaql 124-130 heme oxygenase 1 Homo sapiens 71-75 17600318-8 2007 In functional studies, the HO-1 inhibitor, ZnPP-IX, partly reversed the growth-inhibitory effects of BMP4, and overexpression of HO-1 in PASMCs inhibited serum-stimulated [3H]-thymidine incorporation. zinc protoporphyrin 43-50 heme oxygenase 1 Homo sapiens 27-31 17991645-0 2007 Microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with iron status in persons with type 2 diabetes mellitus. Iron 85-89 heme oxygenase 1 Homo sapiens 35-51 17991645-3 2007 Heme oxygenase (HO) 1 expression is increased when intracellular iron increases. Iron 65-69 heme oxygenase 1 Homo sapiens 0-21 17600318-8 2007 In functional studies, the HO-1 inhibitor, ZnPP-IX, partly reversed the growth-inhibitory effects of BMP4, and overexpression of HO-1 in PASMCs inhibited serum-stimulated [3H]-thymidine incorporation. pasmcs 137-143 heme oxygenase 1 Homo sapiens 129-133 17600318-8 2007 In functional studies, the HO-1 inhibitor, ZnPP-IX, partly reversed the growth-inhibitory effects of BMP4, and overexpression of HO-1 in PASMCs inhibited serum-stimulated [3H]-thymidine incorporation. Tritium 172-174 heme oxygenase 1 Homo sapiens 27-31 17600318-8 2007 In functional studies, the HO-1 inhibitor, ZnPP-IX, partly reversed the growth-inhibitory effects of BMP4, and overexpression of HO-1 in PASMCs inhibited serum-stimulated [3H]-thymidine incorporation. Tritium 172-174 heme oxygenase 1 Homo sapiens 129-133 17600318-8 2007 In functional studies, the HO-1 inhibitor, ZnPP-IX, partly reversed the growth-inhibitory effects of BMP4, and overexpression of HO-1 in PASMCs inhibited serum-stimulated [3H]-thymidine incorporation. Thymidine 176-185 heme oxygenase 1 Homo sapiens 129-133 17868064-10 2007 Diannexin increased expression of HO-1/Bcl-2/Bcl-xl, and reduced Caspase-3/TUNEL+ apoptotic cells. diannexin 0-9 heme oxygenase 1 Homo sapiens 34-38 17600318-10 2007 Inhibition of HO-1 function or expression will further increase the proproliferative capacity of BMPR-II-deficient PASMCs and may thus represent a potential "second hit" necessary for disease manifestation. pasmcs 115-121 heme oxygenase 1 Homo sapiens 14-18 18087801-2 2007 One synthetic derivative of butein, 2",4",6"-tris(methoxymethoxy)chalcone (TMMC), has potent anti-inflammatory activity via an HO-1 (heme oxygenase 1) dependent pathway. butein, 2",4",6"-tris(methoxymethoxy)chalcone 28-73 heme oxygenase 1 Homo sapiens 127-131 17963960-0 2007 The induction of heme oxygenase-1 modulates bismuth oxide-induced cytotoxicity in human dental pulp cells. bismuth oxide 44-57 heme oxygenase 1 Homo sapiens 17-33 17963960-2 2007 We also assessed whether heme oxygenase-1 (HO-1) is involved in BPC-induced cytotoxicity in dental pulp cells. S-(4-bromophenyl)cysteine sulfoxide 64-67 heme oxygenase 1 Homo sapiens 25-41 17963960-2 2007 We also assessed whether heme oxygenase-1 (HO-1) is involved in BPC-induced cytotoxicity in dental pulp cells. S-(4-bromophenyl)cysteine sulfoxide 64-67 heme oxygenase 1 Homo sapiens 43-47 17963960-7 2007 Moreover, BPC-induced HO-1 expression in dental pulp cells plays a protective role against the cytotoxic effects of BPC. S-(4-bromophenyl)cysteine sulfoxide 10-13 heme oxygenase 1 Homo sapiens 22-26 17822364-5 2007 CDDO-Im treatment significantly induced Nrf2-dependent antioxidative genes (HO-1, GCLC, GCLM, and NQO1) in PBMCs isolated from six normal subjects. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 0-7 heme oxygenase 1 Homo sapiens 76-80 17956297-1 2007 The well-known adverse effects of CO (carbon monoxide) intoxication are counterbalanced by its positive actions when small amounts are produced intracellularly by the cytoprotective enzyme HO-1 (haem oxygenase-1). Carbon Monoxide 34-54 heme oxygenase 1 Homo sapiens 189-211 18087801-2 2007 One synthetic derivative of butein, 2",4",6"-tris(methoxymethoxy)chalcone (TMMC), has potent anti-inflammatory activity via an HO-1 (heme oxygenase 1) dependent pathway. butein, 2",4",6"-tris(methoxymethoxy)chalcone 28-73 heme oxygenase 1 Homo sapiens 133-149 18087801-2 2007 One synthetic derivative of butein, 2",4",6"-tris(methoxymethoxy)chalcone (TMMC), has potent anti-inflammatory activity via an HO-1 (heme oxygenase 1) dependent pathway. 2',4',6'-tris(methoxymethoxy) chalcone 75-79 heme oxygenase 1 Homo sapiens 127-131 18087801-2 2007 One synthetic derivative of butein, 2",4",6"-tris(methoxymethoxy)chalcone (TMMC), has potent anti-inflammatory activity via an HO-1 (heme oxygenase 1) dependent pathway. 2',4',6'-tris(methoxymethoxy) chalcone 75-79 heme oxygenase 1 Homo sapiens 133-149 17889572-8 2007 The level of iNOS and HO-1 mRNA expression were increased by CLP, which was prevented by both AG and L-NAME. NG-Nitroarginine Methyl Ester 101-107 heme oxygenase 1 Homo sapiens 22-26 17823375-0 2007 Carbon monoxide induces heme oxygenase-1 via activation of protein kinase R-like endoplasmic reticulum kinase and inhibits endothelial cell apoptosis triggered by endoplasmic reticulum stress. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 24-40 17996129-9 2007 iNOS, COX-2 and HO-1 mRNA expression were fluctuated in the normal range following oligomycin administration (all P>0.05). Oligomycins 83-93 heme oxygenase 1 Homo sapiens 16-20 17915953-1 2007 Heme oxygenase-1 (HO-1) is the chief regulatory enzyme in the oxidative degradation of heme to biliverdin. Heme 87-91 heme oxygenase 1 Homo sapiens 0-16 17915953-1 2007 Heme oxygenase-1 (HO-1) is the chief regulatory enzyme in the oxidative degradation of heme to biliverdin. Heme 87-91 heme oxygenase 1 Homo sapiens 18-22 17915953-1 2007 Heme oxygenase-1 (HO-1) is the chief regulatory enzyme in the oxidative degradation of heme to biliverdin. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 0-16 17915953-1 2007 Heme oxygenase-1 (HO-1) is the chief regulatory enzyme in the oxidative degradation of heme to biliverdin. Biliverdine 95-105 heme oxygenase 1 Homo sapiens 18-22 17915953-2 2007 In the process of heme degradation, HO-1 receives the electrons necessary for catalysis from the flavoprotein NADPH cytochrome P450 reductase (CPR), releasing free iron and carbon monoxide. Iron 164-168 heme oxygenase 1 Homo sapiens 36-40 17915953-2 2007 In the process of heme degradation, HO-1 receives the electrons necessary for catalysis from the flavoprotein NADPH cytochrome P450 reductase (CPR), releasing free iron and carbon monoxide. Carbon Monoxide 173-188 heme oxygenase 1 Homo sapiens 36-40 17915953-10 2007 The R254K full-length form had a specific activity of approximately 200-225 nmol of bilirubin h-1 nmol-1 HO-1 as compared to approximately 140-150 nmol of bilirubin h-1 nmol-1 for the WT form, which contains the 30 kDa contaminant. Bilirubin 84-93 heme oxygenase 1 Homo sapiens 105-109 17915953-10 2007 The R254K full-length form had a specific activity of approximately 200-225 nmol of bilirubin h-1 nmol-1 HO-1 as compared to approximately 140-150 nmol of bilirubin h-1 nmol-1 for the WT form, which contains the 30 kDa contaminant. bilirubin h 84-95 heme oxygenase 1 Homo sapiens 105-109 17915953-12 2007 Because the membrane-spanning domain is present, the full-length hHO-1 has the potential to incorporate into phospholipid membranes, which can be reconstituted at known concentrations, in combination with other endoplasmic reticulum resident enzymes. Phospholipids 109-121 heme oxygenase 1 Homo sapiens 65-70 17823375-1 2007 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, is antiapoptotic in a variety of models of cellular injury, but the precise mechanisms remain to be established. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 63-84 17823375-1 2007 Carbon monoxide (CO), a reaction product of the cytoprotective heme oxygenase (HO)-1, is antiapoptotic in a variety of models of cellular injury, but the precise mechanisms remain to be established. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 63-84 17895889-6 2007 Cobalt protoporphyrin induced HMOX-1, instead of HIF-1 alpha expression and increased bortezomib-induced apoptosis in the presence of pharmacologically effective doses of bortezomib. cobaltiprotoporphyrin 0-21 heme oxygenase 1 Homo sapiens 30-36 17967796-0 2007 A new road to induce heme oxygenase-1 expression by carbon monoxide. Carbon Monoxide 52-67 heme oxygenase 1 Homo sapiens 21-37 17895889-7 2007 In contrast, zinc protoporphyrin downregulated HMOX-1 expression, thereby partially inhibiting bortezomib-induced cell death. zinc protoporphyrin 13-32 heme oxygenase 1 Homo sapiens 47-53 17895889-0 2007 Induction of heme oxygenase-1 by cobalt protoporphyrin enhances the antitumour effect of bortezomib in adult T-cell leukaemia cells. cobaltiprotoporphyrin 33-54 heme oxygenase 1 Homo sapiens 13-29 17895889-7 2007 In contrast, zinc protoporphyrin downregulated HMOX-1 expression, thereby partially inhibiting bortezomib-induced cell death. Bortezomib 95-105 heme oxygenase 1 Homo sapiens 47-53 17895889-0 2007 Induction of heme oxygenase-1 by cobalt protoporphyrin enhances the antitumour effect of bortezomib in adult T-cell leukaemia cells. Bortezomib 89-99 heme oxygenase 1 Homo sapiens 13-29 17895889-8 2007 This indicates that HMOX-1 may modulate anticancer effects of bortezomib in ATL cells, and could be a molecular target in treating ATL patients. Bortezomib 62-72 heme oxygenase 1 Homo sapiens 20-26 17895889-5 2007 An oligonucleotide DNA microarray analysis of TaY cells revealed upregulation of genes encoding heat shock proteins (HSPA1A, STIP1, HSPA1B, and HSPCA), genes related to protein folding (CDC37 and ANAPC5), Fas-associated factor 1(FAF1) and an oxidative stress-related gene, heme oxygenase-1(HMOX-1), known to be a target gene of hypoxia-inducible gene-1 alpha (HIF-1 alpha). Oligonucleotides 3-18 heme oxygenase 1 Homo sapiens 273-289 17895889-5 2007 An oligonucleotide DNA microarray analysis of TaY cells revealed upregulation of genes encoding heat shock proteins (HSPA1A, STIP1, HSPA1B, and HSPCA), genes related to protein folding (CDC37 and ANAPC5), Fas-associated factor 1(FAF1) and an oxidative stress-related gene, heme oxygenase-1(HMOX-1), known to be a target gene of hypoxia-inducible gene-1 alpha (HIF-1 alpha). Oligonucleotides 3-18 heme oxygenase 1 Homo sapiens 290-296 17896171-8 2007 RESULTS: HO-1-overexpression reduced proliferative rates and DNA-synthesis of HUVEC, but provided potent protection from oxidative stress induced by heme and H(2)O(2). Heme 149-153 heme oxygenase 1 Homo sapiens 9-13 17919491-1 2007 BACKGROUND & AIMS: Induction of heme oxygenase-1 (HO-1) has been shown to be beneficial in immune-mediated liver damage. Adenosine Monophosphate 12-15 heme oxygenase 1 Homo sapiens 36-52 17919491-1 2007 BACKGROUND & AIMS: Induction of heme oxygenase-1 (HO-1) has been shown to be beneficial in immune-mediated liver damage. Adenosine Monophosphate 12-15 heme oxygenase 1 Homo sapiens 54-58 17919491-7 2007 HO-1 was induced by either cobalt-protoporphyrin-IX or over expressed by adenoviral gene transfer. cobaltiprotoporphyrin 27-51 heme oxygenase 1 Homo sapiens 0-4 17919492-1 2007 BACKGROUND & AIMS: Heme oxygenase-1 (HO-1) is an antioxidant defense and key cytoprotective enzyme, which is repressed by Bach1. Adenosine Monophosphate 12-15 heme oxygenase 1 Homo sapiens 23-39 17919492-1 2007 BACKGROUND & AIMS: Heme oxygenase-1 (HO-1) is an antioxidant defense and key cytoprotective enzyme, which is repressed by Bach1. Adenosine Monophosphate 12-15 heme oxygenase 1 Homo sapiens 41-45 17919492-3 2007 This study was to assess whether a specific miR-122 antagomir down-regulates HCV protein replication and up-regulates HO-1. mir-122 44-51 heme oxygenase 1 Homo sapiens 118-122 17919492-7 2007 Antagomir of miR-122 also decreased Bach1 and increased HO-1 mRNA levels in CNS3, 9-13, and WT Huh-7 cells. mir-122 13-20 heme oxygenase 1 Homo sapiens 56-60 17919492-8 2007 Increasing HO-1 by silencing Bach1 with 50 nmol/L Bach1-short interfering RNA or by treatment with 5 mumol/L cobalt protoporphyrin or heme (known inducers of HO-1) decreased HCV RNA and protein by 50% in HCV replicon cells. cobaltiprotoporphyrin 109-130 heme oxygenase 1 Homo sapiens 11-15 17919492-8 2007 Increasing HO-1 by silencing Bach1 with 50 nmol/L Bach1-short interfering RNA or by treatment with 5 mumol/L cobalt protoporphyrin or heme (known inducers of HO-1) decreased HCV RNA and protein by 50% in HCV replicon cells. cobaltiprotoporphyrin 109-130 heme oxygenase 1 Homo sapiens 158-162 17919492-8 2007 Increasing HO-1 by silencing Bach1 with 50 nmol/L Bach1-short interfering RNA or by treatment with 5 mumol/L cobalt protoporphyrin or heme (known inducers of HO-1) decreased HCV RNA and protein by 50% in HCV replicon cells. Heme 134-138 heme oxygenase 1 Homo sapiens 11-15 17919492-8 2007 Increasing HO-1 by silencing Bach1 with 50 nmol/L Bach1-short interfering RNA or by treatment with 5 mumol/L cobalt protoporphyrin or heme (known inducers of HO-1) decreased HCV RNA and protein by 50% in HCV replicon cells. Heme 134-138 heme oxygenase 1 Homo sapiens 158-162 17919492-10 2007 Increasing HO-1, by silencing the Bach1 gene or by treatment with cobalt protoporphyrin or heme, decreases HCV replication. cobaltiprotoporphyrin 66-87 heme oxygenase 1 Homo sapiens 11-15 17679649-1 2007 Our previous studies suggest that heme oxygenase (HO)-1 induction and/or subsequent bilirubin generation in endothelial cells may suppress superoxide generation of from reduced nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase. Superoxides 139-149 heme oxygenase 1 Homo sapiens 34-55 17919492-10 2007 Increasing HO-1, by silencing the Bach1 gene or by treatment with cobalt protoporphyrin or heme, decreases HCV replication. Heme 91-95 heme oxygenase 1 Homo sapiens 11-15 17679649-5 2007 The increase in HO-1 activity and inhibition of NADPH oxidase activity by hemin were reversed by tin protoporphyrin-IX and were not associated with changes in Nox2 or Nox4 protein levels. tin protoporphyrin IX 97-118 heme oxygenase 1 Homo sapiens 16-20 17919492-11 2007 Thus, miR-122 plays an important role in the regulation of HCV replication and HO-1/Bach1 expression in hepatocytes. mir-122 6-13 heme oxygenase 1 Homo sapiens 79-83 17679649-10 2007 Thus, systemic expression of HO-1 suppresses NADPH oxidase activity by mechanisms at least partly mediated by the bile pigment bilirubin, thereby reducing oxidative stress. Bilirubin 127-136 heme oxygenase 1 Homo sapiens 29-33 18044280-1 2007 INTRODUCTION: Heme-Oxygenase-1 catalyzes hemoglobin into bilirubin, iron, and carbon monoxide, a well known vasodilator. Bilirubin 57-66 heme oxygenase 1 Homo sapiens 14-30 18044280-1 2007 INTRODUCTION: Heme-Oxygenase-1 catalyzes hemoglobin into bilirubin, iron, and carbon monoxide, a well known vasodilator. Iron 68-72 heme oxygenase 1 Homo sapiens 14-30 18044280-1 2007 INTRODUCTION: Heme-Oxygenase-1 catalyzes hemoglobin into bilirubin, iron, and carbon monoxide, a well known vasodilator. Carbon Monoxide 78-93 heme oxygenase 1 Homo sapiens 14-30 17678632-0 2007 2",4",6"-tris(methoxymethoxy) chalcone protects against trinitrobenzene sulfonic acid-induced colitis and blocks tumor necrosis factor-alpha-induced intestinal epithelial inflammation via heme oxygenase 1-dependent and independent pathways. 2',4',6'-tris(methoxymethoxy) chalcone 0-38 heme oxygenase 1 Homo sapiens 188-204 17678632-6 2007 TMMC induced the expression of heme oxygenase 1 (HO-1) in HT-29 cells. 2',4',6'-tris(methoxymethoxy) chalcone 0-4 heme oxygenase 1 Homo sapiens 31-47 17678632-6 2007 TMMC induced the expression of heme oxygenase 1 (HO-1) in HT-29 cells. 2',4',6'-tris(methoxymethoxy) chalcone 0-4 heme oxygenase 1 Homo sapiens 49-53 17678632-7 2007 TMMC increased extracellular signal-regulated kinase1/2 and p38 kinase phosphorylation levels, which led to the nuclear translocation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and consequently to HO-1 expression. 2',4',6'-tris(methoxymethoxy) chalcone 0-4 heme oxygenase 1 Homo sapiens 208-212 17678632-10 2007 Moreover, we found that the different products of HO-1, carbon monoxide and bilirubin, exerted anti-inflammatory effects that were additive or synergistic in HT-29 cells stimulated with TNF-alpha. Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 50-54 17678632-10 2007 Moreover, we found that the different products of HO-1, carbon monoxide and bilirubin, exerted anti-inflammatory effects that were additive or synergistic in HT-29 cells stimulated with TNF-alpha. Bilirubin 76-85 heme oxygenase 1 Homo sapiens 50-54 17726138-0 2007 Genetic variability in iron-related oxidative stress pathways (Nrf2, NQ01, NOS3, and HO-1), iron intake, and risk of postmenopausal breast cancer. Iron 23-27 heme oxygenase 1 Homo sapiens 85-89 17567933-3 2007 HO-1 catabolizes pro-oxidant heme into substances with anti-oxidant, anti-apoptotic, anti-fibrogenic, vasodilatory and immune modulatory properties. Heme 29-33 heme oxygenase 1 Homo sapiens 0-4 17567933-7 2007 Actinomycin D and nuclear run-on studies demonstrate that TGF-beta1 augments HO-1 expression by increased gene transcription and does not involve increased mRNA stability. Dactinomycin 0-13 heme oxygenase 1 Homo sapiens 77-81 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Heme 139-143 heme oxygenase 1 Homo sapiens 28-32 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 28-32 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Carbon Monoxide 166-168 heme oxygenase 1 Homo sapiens 28-32 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Iron 171-175 heme oxygenase 1 Homo sapiens 28-32 17729116-0 2007 Hemin-induced Erk1/2 activation and heme oxygenase-1 expression in human umbilical vein endothelial cells. Hemin 0-5 heme oxygenase 1 Homo sapiens 36-52 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Iron 177-179 heme oxygenase 1 Homo sapiens 28-32 17569621-7 2007 Upon organ transplantation, HO-1 is ubiquitously expressed in a transplanted organ, becoming the rate-limiting enzyme in the catabolism of heme into carbon monoxide (CO), iron (Fe) and biliverdin (1). Biliverdine 185-195 heme oxygenase 1 Homo sapiens 28-32 17418561-0 2007 Piperine protects cisplatin-induced apoptosis via heme oxygenase-1 induction in auditory cells. piperine 0-8 heme oxygenase 1 Homo sapiens 50-66 17917164-1 2007 In this study, we evaluated the effect of lipoic acid (LA) and N-acetyl cysteine (NAC) on oxidative [4-hydroxy-2-nonenal, N(epsilon)-(carboxymethyl)lysine and heme oxygenase-1] and apoptotic (caspase 9 and Bax) markers in fibroblasts from patients with Alzheimer disease (AD) and age-matched and young controls. Thioctic Acid 42-53 heme oxygenase 1 Homo sapiens 159-175 17917164-1 2007 In this study, we evaluated the effect of lipoic acid (LA) and N-acetyl cysteine (NAC) on oxidative [4-hydroxy-2-nonenal, N(epsilon)-(carboxymethyl)lysine and heme oxygenase-1] and apoptotic (caspase 9 and Bax) markers in fibroblasts from patients with Alzheimer disease (AD) and age-matched and young controls. Acetylcysteine 82-85 heme oxygenase 1 Homo sapiens 159-175 17418561-0 2007 Piperine protects cisplatin-induced apoptosis via heme oxygenase-1 induction in auditory cells. Cisplatin 18-27 heme oxygenase 1 Homo sapiens 50-66 17418561-2 2007 In this study, we examined whether piperine could protect House Ear Institute-Organ of Corti 1 (HEI-OC1) cells against cisplatin-induced apoptosis through the induction of heme oxygenase (HO)-1 expression. piperine 35-43 heme oxygenase 1 Homo sapiens 172-193 17573819-9 2007 Stress-induced up-regulation of the heme-degrading enzyme, heme oxygenase-1 in AD-affected astroglia may impact central lipid homeostasis by promoting the oxidation of cholesterol to a host of oxysterol intermediates. Heme 36-40 heme oxygenase 1 Homo sapiens 59-75 17418561-3 2007 Piperine (10-100 microM) induced the expression of HO-1 in dose- and time-dependent manners. piperine 0-8 heme oxygenase 1 Homo sapiens 51-55 17573819-9 2007 Stress-induced up-regulation of the heme-degrading enzyme, heme oxygenase-1 in AD-affected astroglia may impact central lipid homeostasis by promoting the oxidation of cholesterol to a host of oxysterol intermediates. Cholesterol 168-179 heme oxygenase 1 Homo sapiens 59-75 17418561-5 2007 Piperine activated the c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase and p38 mitogen-activated protein kinase (MAPK) pathways, and the JNK pathway played an important role in piperine-induced HO-1 expression. piperine 0-8 heme oxygenase 1 Homo sapiens 215-219 17573819-9 2007 Stress-induced up-regulation of the heme-degrading enzyme, heme oxygenase-1 in AD-affected astroglia may impact central lipid homeostasis by promoting the oxidation of cholesterol to a host of oxysterol intermediates. Oxysterols 193-202 heme oxygenase 1 Homo sapiens 59-75 17418561-5 2007 Piperine activated the c-Jun N-terminal kinase (JNK), extracellular signal-regulated kinase and p38 mitogen-activated protein kinase (MAPK) pathways, and the JNK pathway played an important role in piperine-induced HO-1 expression. piperine 198-206 heme oxygenase 1 Homo sapiens 215-219 17418561-7 2007 The protective effect of piperine was abrogated by zinc protoporphyrin IX, an HO inhibitor, and antisense oligodeoxynucleotides against HO-1 gene. piperine 25-33 heme oxygenase 1 Homo sapiens 136-140 17418561-8 2007 These results demonstrate that the expression of HO-1 by piperine is mediated by both JNK pathway and Nrf2, and the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells. piperine 57-65 heme oxygenase 1 Homo sapiens 49-53 17418561-8 2007 These results demonstrate that the expression of HO-1 by piperine is mediated by both JNK pathway and Nrf2, and the expression inhibits cisplatin-induced apoptosis in HEI-OC1 cells. Cisplatin 136-145 heme oxygenase 1 Homo sapiens 49-53 17785948-9 2007 Accordingly, under the heme-rich condition, heme binds to cysteine-proline (CP) motifs of ALAS1 and those of transcriptional repressor Bach1, thereby leading to repression of mitochondrial transport of ALAS1 and induction of HO-1 transcription, respectively. Heme 44-48 heme oxygenase 1 Homo sapiens 225-229 17675106-1 2007 We investigated whether increased heme oxygenase (HO)-1 activity by NS-398 is responsible for protection against hypoxia-induced damage in C6 cells. Nitrogen 68-70 heme oxygenase 1 Homo sapiens 34-55 17675106-3 2007 NS-398 increased HO-1 expression in a concentration- and time-dependent manner during both normoxia and hypoxia (95% N(2)/5% CO(2)), but the latter was much more sensitive. Nitrogen 0-2 heme oxygenase 1 Homo sapiens 17-21 17675106-3 2007 NS-398 increased HO-1 expression in a concentration- and time-dependent manner during both normoxia and hypoxia (95% N(2)/5% CO(2)), but the latter was much more sensitive. Nitrogen 117-121 heme oxygenase 1 Homo sapiens 17-21 17675106-3 2007 NS-398 increased HO-1 expression in a concentration- and time-dependent manner during both normoxia and hypoxia (95% N(2)/5% CO(2)), but the latter was much more sensitive. co(2)) 125-131 heme oxygenase 1 Homo sapiens 17-21 17675106-4 2007 Because induction of HO-1 occurred due to hypoxia itself, NS-398 seemed to potentiate the expression of HO-1. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 58-64 heme oxygenase 1 Homo sapiens 21-25 17675106-4 2007 Because induction of HO-1 occurred due to hypoxia itself, NS-398 seemed to potentiate the expression of HO-1. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 58-64 heme oxygenase 1 Homo sapiens 104-108 17675106-6 2007 Zinc protophorphrin (ZnPPIX), a HO-1 inhibitor, inhibited the protective effect of NS-398 against hypoxia. zinc protophorphrin 0-19 heme oxygenase 1 Homo sapiens 32-36 17675106-6 2007 Zinc protophorphrin (ZnPPIX), a HO-1 inhibitor, inhibited the protective effect of NS-398 against hypoxia. zinc protoporphyrin 21-27 heme oxygenase 1 Homo sapiens 32-36 17675106-6 2007 Zinc protophorphrin (ZnPPIX), a HO-1 inhibitor, inhibited the protective effect of NS-398 against hypoxia. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 83-89 heme oxygenase 1 Homo sapiens 32-36 17675106-10 2007 NS-398 increased phosphorylation of Akt, and LY-294002, a specific PI(3) kinase inhibitor, inhibited NS-398-induced HO-1 expression. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 0-6 heme oxygenase 1 Homo sapiens 116-120 17675106-10 2007 NS-398 increased phosphorylation of Akt, and LY-294002, a specific PI(3) kinase inhibitor, inhibited NS-398-induced HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 45-54 heme oxygenase 1 Homo sapiens 116-120 17675106-10 2007 NS-398 increased phosphorylation of Akt, and LY-294002, a specific PI(3) kinase inhibitor, inhibited NS-398-induced HO-1 expression. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 101-107 heme oxygenase 1 Homo sapiens 116-120 17675106-11 2007 Taken together, we conclude that therapeutic use of NS-398 in the treatment of oxidative stress-oriented neuronal disorders would be beneficial through dual actions: HO-1 induction and COX-2 inhibition. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 52-58 heme oxygenase 1 Homo sapiens 166-170 17785948-7 2007 Heme reduces heme synthesis by suppressing the expression of non-specific 5-aminolevulinate synthase (ALAS1) and stimulates heme breakdown by inducing heme oxygenase (HO)-1 expression. Heme 0-4 heme oxygenase 1 Homo sapiens 151-172 17785948-8 2007 ALAS1 and HO-1 are the rate limiting enzymes in heme biosynthesis and catabolism, respectively. Heme 48-52 heme oxygenase 1 Homo sapiens 10-14 17785948-9 2007 Accordingly, under the heme-rich condition, heme binds to cysteine-proline (CP) motifs of ALAS1 and those of transcriptional repressor Bach1, thereby leading to repression of mitochondrial transport of ALAS1 and induction of HO-1 transcription, respectively. Heme 23-27 heme oxygenase 1 Homo sapiens 225-229 17785948-9 2007 Accordingly, under the heme-rich condition, heme binds to cysteine-proline (CP) motifs of ALAS1 and those of transcriptional repressor Bach1, thereby leading to repression of mitochondrial transport of ALAS1 and induction of HO-1 transcription, respectively. cysteine-proline 58-74 heme oxygenase 1 Homo sapiens 225-229 17665394-2 2007 METHODS: In the K/BxN mouse serum transfer model, which mimics human inflammatory arthritis without lymphocyte influence, HO-1 was up-regulated by intraperitoneal injection of cobalt protoporphyrin IX (CoPP), a potent pharmacologic inducer, and was inhibited using a specific siRNA. cobaltiprotoporphyrin 176-200 heme oxygenase 1 Homo sapiens 122-126 17520317-2 2007 A (GT)n dinucleotide repeat in HO-1 promoter is polymorphic and modulates the transcriptional activity of the gene. Dinucleoside Phosphates 8-20 heme oxygenase 1 Homo sapiens 31-35 17665394-2 2007 METHODS: In the K/BxN mouse serum transfer model, which mimics human inflammatory arthritis without lymphocyte influence, HO-1 was up-regulated by intraperitoneal injection of cobalt protoporphyrin IX (CoPP), a potent pharmacologic inducer, and was inhibited using a specific siRNA. cobaltiprotoporphyrin 202-206 heme oxygenase 1 Homo sapiens 122-126 17433488-0 2007 Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. Quercetin 0-9 heme oxygenase 1 Homo sapiens 87-103 17671894-14 2007 On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 17-21 17671894-14 2007 On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Biliverdine 120-130 heme oxygenase 1 Homo sapiens 105-109 17671894-14 2007 On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Bilirubin 157-166 heme oxygenase 1 Homo sapiens 17-21 17671894-14 2007 On the contrary, HO-1 is highly upregulated and it plays a very important role of antioxidation, because HO-1 generates biliverdin, which being converted to bilirubin exhibits a very potent antioxidative effect, and hence antiapoptosis in tumors. Bilirubin 157-166 heme oxygenase 1 Homo sapiens 105-109 17671894-15 2007 Thus this oxidation therapy, by inhibiting this HO-1 dependent antioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeutic modality for future anticancer therapy. Bilirubin 76-85 heme oxygenase 1 Homo sapiens 48-52 17295091-0 2007 Heme oxygenase-1 and interleukin-11 are overexpressed in stress-induced premature senescence of human WI-38 fibroblasts induced by tert-butylhydroperoxide and ethanol. tert-Butylhydroperoxide 131-154 heme oxygenase 1 Homo sapiens 0-16 17295091-0 2007 Heme oxygenase-1 and interleukin-11 are overexpressed in stress-induced premature senescence of human WI-38 fibroblasts induced by tert-butylhydroperoxide and ethanol. Ethanol 159-166 heme oxygenase 1 Homo sapiens 0-16 17295091-2 2007 In the present work we found an increased mRNA and protein level of interleukin-11 and heme oxygenase-1 in premature senescence of WI-38 human diploid foetal lung fibroblasts induced by both tert-butylhydroperoxide and ethanol. tert-Butylhydroperoxide 191-214 heme oxygenase 1 Homo sapiens 87-103 17295091-2 2007 In the present work we found an increased mRNA and protein level of interleukin-11 and heme oxygenase-1 in premature senescence of WI-38 human diploid foetal lung fibroblasts induced by both tert-butylhydroperoxide and ethanol. Ethanol 219-226 heme oxygenase 1 Homo sapiens 87-103 17295091-3 2007 We tested whether interleukin-11 and heme oxygenase-1 could protect against tert-butylhydroperoxide- or ethanol-induced premature senescence when stable overexpression was established using a retroviral vector-based transduction. tert-Butylhydroperoxide 76-99 heme oxygenase 1 Homo sapiens 37-53 17295091-3 2007 We tested whether interleukin-11 and heme oxygenase-1 could protect against tert-butylhydroperoxide- or ethanol-induced premature senescence when stable overexpression was established using a retroviral vector-based transduction. Ethanol 104-111 heme oxygenase 1 Homo sapiens 37-53 17701549-2 2007 The transcription of Hmox-1 is regulated by the substrate of HO-1, heme. Heme 67-71 heme oxygenase 1 Homo sapiens 21-27 17701549-2 2007 The transcription of Hmox-1 is regulated by the substrate of HO-1, heme. Heme 67-71 heme oxygenase 1 Homo sapiens 61-65 17701549-3 2007 Heme induces expression of Hmox-1 in part by inhibiting the binding of Bach1 to the enhancers and inducing the nuclear export of Bach1. Heme 0-4 heme oxygenase 1 Homo sapiens 27-33 17671894-15 2007 Thus this oxidation therapy, by inhibiting this HO-1 dependent antioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeutic modality for future anticancer therapy. zinc protoporphyrin 100-104 heme oxygenase 1 Homo sapiens 48-52 17671894-15 2007 Thus this oxidation therapy, by inhibiting this HO-1 dependent antioxidant (bilirubin) formation by ZnPP, and by enhancing ROS generation, is expected to offer a powerful therapeutic modality for future anticancer therapy. Reactive Oxygen Species 123-126 heme oxygenase 1 Homo sapiens 48-52 17433488-0 2007 Quercetin protects human hepatocytes from ethanol-derived oxidative stress by inducing heme oxygenase-1 via the MAPK/Nrf2 pathways. Ethanol 42-49 heme oxygenase 1 Homo sapiens 87-103 17673168-0 2007 WITHDRAWN: Heme oxygenase-1 mediates the anti-inflammatory effect of propyl gallate in LPS-stimulated macrophages. Propyl Gallate 69-83 heme oxygenase 1 Homo sapiens 11-27 17559630-6 2007 CONCLUSION: These data suggest that heme oxygenase-1 induction plays a protective role in periodontal ligament cells against the cytotoxic and RANKL-inducing effects of H2O2, through multiple signaling pathways. Hydrogen Peroxide 169-173 heme oxygenase 1 Homo sapiens 36-52 17559630-0 2007 Defense mechanism of heme oxygenase-1 against cytotoxic and receptor activator of nuclear factor-kappaB ligand inducing effects of hydrogen peroxide in human periodontal ligament cells. Hydrogen Peroxide 131-148 heme oxygenase 1 Homo sapiens 21-37 17559630-1 2007 BACKGROUND AND OBJECTIVE: Although induction of heme oxygenase-1 by H2O2 has been reported, the protective role of heme oxygenase-1 against the cytotoxic and osteoclastogenic effects of H2O2 have not been elucidated in human periodontal ligament cells. Hydrogen Peroxide 68-72 heme oxygenase 1 Homo sapiens 48-64 17559630-1 2007 BACKGROUND AND OBJECTIVE: Although induction of heme oxygenase-1 by H2O2 has been reported, the protective role of heme oxygenase-1 against the cytotoxic and osteoclastogenic effects of H2O2 have not been elucidated in human periodontal ligament cells. Hydrogen Peroxide 186-190 heme oxygenase 1 Homo sapiens 115-131 17559630-2 2007 The aim of this work was to investigate the defense mechanism of heme oxygenase-1 on H2O2-induced cytotoxicity and to analyze the expression of receptor activator of nuclear factor-kappaB ligand (RANKL) and osteoprotegerin as markers for osteoclast differentiation in periodontal ligament cells. Hydrogen Peroxide 85-89 heme oxygenase 1 Homo sapiens 65-81 17559630-4 2007 RESULTS: H2O2 produced a cytotoxic effect by reducing the cell viability and enhancing the expression of heme oxygenase-1 and RANKL mRNAs in a concentration- and time-dependent manner. Hydrogen Peroxide 9-13 heme oxygenase 1 Homo sapiens 105-121 17526500-9 2007 The endogenous expression of HO-1 through ER stress-initiated ROS is believed to be as a protection signal. Reactive Oxygen Species 62-65 heme oxygenase 1 Homo sapiens 29-33 17430897-1 2007 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is an integral membrane protein of the smooth endoplasmic reticulum. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 17420286-8 2007 Moreover, 2 Hsp32-targeting drugs, pegylated zinc protoporphyrin (PEG-ZnPP) and styrene maleic acid copolymer micelle-encapsulated ZnPP (SMA-ZnPP), were found to inhibit proliferation and to induce apoptosis in neoplastic MCs. styrene maleic acid copolymer 80-109 heme oxygenase 1 Homo sapiens 12-17 17420286-8 2007 Moreover, 2 Hsp32-targeting drugs, pegylated zinc protoporphyrin (PEG-ZnPP) and styrene maleic acid copolymer micelle-encapsulated ZnPP (SMA-ZnPP), were found to inhibit proliferation and to induce apoptosis in neoplastic MCs. copoly(styrene-maleic acid)-zinc protoporphyrin 137-145 heme oxygenase 1 Homo sapiens 12-17 17502383-9 2007 Finally, heme induced oxidative burst, neutrophil recruitment, and heme oxygenase-1 expression independently of TLR4. Heme 9-13 heme oxygenase 1 Homo sapiens 67-83 17430897-1 2007 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is an integral membrane protein of the smooth endoplasmic reticulum. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 17430897-2 2007 However, we detected an HO-1 immunoreactive signal in the nucleus of cultured cells after exposure to hypoxia and heme or heme/hemopexin. Heme 114-118 heme oxygenase 1 Homo sapiens 24-28 17430897-2 2007 However, we detected an HO-1 immunoreactive signal in the nucleus of cultured cells after exposure to hypoxia and heme or heme/hemopexin. Heme 122-126 heme oxygenase 1 Homo sapiens 24-28 17430897-5 2007 Incubation with leptomycin B prior to hypoxia abolished nuclear HO-1 and the faster migrating band on Western analysis, suggesting that this process was facilitated by CRM1. leptomycin B 16-28 heme oxygenase 1 Homo sapiens 64-68 17430897-9 2007 Nevertheless, nuclear HO-1 protected cells against hydrogen peroxide-mediated injury equally as well as cytoplasmic HO-1. Hydrogen Peroxide 51-68 heme oxygenase 1 Homo sapiens 22-26 17508911-7 2007 These novel findings provide a link between the increase in HO-1 activity, with its concurrent enhanced production of carbon monoxide (CO) and bilirubin, a decrease in infiltrated CD11c(+) dendritic cells and an increase in anti-apoptotic proteins, including RSK and BcL-xL, in the interdiction of the diabetic state. Carbon Monoxide 118-133 heme oxygenase 1 Homo sapiens 60-64 17508911-7 2007 These novel findings provide a link between the increase in HO-1 activity, with its concurrent enhanced production of carbon monoxide (CO) and bilirubin, a decrease in infiltrated CD11c(+) dendritic cells and an increase in anti-apoptotic proteins, including RSK and BcL-xL, in the interdiction of the diabetic state. Carbon Monoxide 135-137 heme oxygenase 1 Homo sapiens 60-64 17508911-7 2007 These novel findings provide a link between the increase in HO-1 activity, with its concurrent enhanced production of carbon monoxide (CO) and bilirubin, a decrease in infiltrated CD11c(+) dendritic cells and an increase in anti-apoptotic proteins, including RSK and BcL-xL, in the interdiction of the diabetic state. Bilirubin 143-152 heme oxygenase 1 Homo sapiens 60-64 17639610-5 2007 The levels of HO-1 showed a significant positive correlated with parameters of renal function, creatinine clearance, and renal blood flow and urine output (AUC; r = 0.8042, P = 0.03; r = 0.6028, P = 0.04; r = 0.6055, P = 0.04), demonstrating a possible protective role of this gene in this model of renal transplantation. Creatinine 95-105 heme oxygenase 1 Homo sapiens 14-18 17627585-2 2007 When free heme concentration is increased, it results in the induction of heme oxygenase-1 (HO-1), which then breaks free heme down. Heme 10-14 heme oxygenase 1 Homo sapiens 74-90 17627585-2 2007 When free heme concentration is increased, it results in the induction of heme oxygenase-1 (HO-1), which then breaks free heme down. Heme 10-14 heme oxygenase 1 Homo sapiens 92-96 17627585-2 2007 When free heme concentration is increased, it results in the induction of heme oxygenase-1 (HO-1), which then breaks free heme down. Heme 74-78 heme oxygenase 1 Homo sapiens 92-96 17349619-5 2007 The mechanisms by which salidroside protected neuron cells from oxidative stress included the induction of several antioxidant enzymes, thioredoxin, heme oxygenase-1, and peroxiredoxin-I; the downregulation of pro-apoptotic gene Bax and the upregulation of anti-apoptotic genes Bcl-2 and Bcl-X(L). rhodioloside 24-35 heme oxygenase 1 Homo sapiens 149-165 17391667-0 2007 The alpha-methylene-gamma-butyrolactone moiety in dehydrocostus lactone is responsible for cytoprotective heme oxygenase-1 expression through activation of the nuclear factor E2-related factor 2 in HepG2 cells. Lactones 32-39 heme oxygenase 1 Homo sapiens 106-122 17603281-7 2007 In human aortic smooth muscle cells (HASMCs), curcumin also inhibited growth triggered by TNF-alpha and increased p21(WAF1/CIP1) expression via HO-1-dependent manner. Curcumin 46-54 heme oxygenase 1 Homo sapiens 144-148 17490617-6 2007 Binding of hemoglobin-haptoglobin complex (Hb/Hp) to CD163 resulted in significant induction of HO-1 by peripheral blood mononuclear cells after exposure to dexamethasone prior to culture. Dexamethasone 157-170 heme oxygenase 1 Homo sapiens 96-100 17490617-9 2007 Steroid treatment was suggested to facilitate CD163-mediated endocytosis of hemoglobin to monocytes/macrophages and thereby induce acceleration of HO-1 synthesis. Steroids 0-7 heme oxygenase 1 Homo sapiens 147-151 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 0-16 17449870-0 2007 Photooxidation generates biologically active phospholipids that induce heme oxygenase-1 in skin cells. Phospholipids 45-58 heme oxygenase 1 Homo sapiens 71-87 17449870-2 2007 HO-1 enzymatic activity results in the formation of the cytoprotective metabolites CO and biliverdin. Carbon Monoxide 83-85 heme oxygenase 1 Homo sapiens 0-4 17449870-2 2007 HO-1 enzymatic activity results in the formation of the cytoprotective metabolites CO and biliverdin. Biliverdine 90-100 heme oxygenase 1 Homo sapiens 0-4 17449870-4 2007 Here we show that UVA-1 irradiation generates oxidized phospholipids derived from 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC) that mediate the expression of HO-1 in skin cells. Phospholipids 55-68 heme oxygenase 1 Homo sapiens 178-182 17449870-4 2007 Here we show that UVA-1 irradiation generates oxidized phospholipids derived from 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC) that mediate the expression of HO-1 in skin cells. 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine 82-139 heme oxygenase 1 Homo sapiens 178-182 17449870-4 2007 Here we show that UVA-1 irradiation generates oxidized phospholipids derived from 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (PAPC) that mediate the expression of HO-1 in skin cells. PAPC 141-145 heme oxygenase 1 Homo sapiens 178-182 17449870-6 2007 Irradiation of arachidonate-containing phospholipids with UVA-1 led to formation of defined lipid oxidation products including epoxyisoprostane-phosphatidylcholine that induced HO-1 expression in dermal fibroblasts, in keratinocytes, and in a three-dimensional epidermal equivalent model. Arachidonic Acid 15-27 heme oxygenase 1 Homo sapiens 177-181 17449870-6 2007 Irradiation of arachidonate-containing phospholipids with UVA-1 led to formation of defined lipid oxidation products including epoxyisoprostane-phosphatidylcholine that induced HO-1 expression in dermal fibroblasts, in keratinocytes, and in a three-dimensional epidermal equivalent model. Phospholipids 39-52 heme oxygenase 1 Homo sapiens 177-181 17449870-6 2007 Irradiation of arachidonate-containing phospholipids with UVA-1 led to formation of defined lipid oxidation products including epoxyisoprostane-phosphatidylcholine that induced HO-1 expression in dermal fibroblasts, in keratinocytes, and in a three-dimensional epidermal equivalent model. epoxyisoprostane-phosphatidylcholine 127-163 heme oxygenase 1 Homo sapiens 177-181 17449870-8 2007 Together, we present a novel mechanism of UVA-1-induced HO-1 expression that is mediated by the generation of biologically active phospholipid oxidation products. Phospholipids 130-142 heme oxygenase 1 Homo sapiens 56-60 17511582-6 2007 After sleep in OSA patients, changes were noted in several genes involved in modulation of reactive oxygen species (ROS), including heme oxygenase 1, superoxide dismutase 1 and 2, and catalase. Reactive Oxygen Species 91-114 heme oxygenase 1 Homo sapiens 132-148 17511582-6 2007 After sleep in OSA patients, changes were noted in several genes involved in modulation of reactive oxygen species (ROS), including heme oxygenase 1, superoxide dismutase 1 and 2, and catalase. Reactive Oxygen Species 116-119 heme oxygenase 1 Homo sapiens 132-148 17095152-0 2007 Functional interaction between nitric oxide-induced iron homeostasis and heme oxygenase-1 in immortalized and malignant oral keratinocytes. Nitric Oxide 31-43 heme oxygenase 1 Homo sapiens 73-89 17095152-10 2007 NO-induced cytotoxicity was also inhibited by hemin (an HO-1 agonist) and was enhanced by zinc protoporphyrin IX (an HO-1 inhibitor). zinc protoporphyrin 90-112 heme oxygenase 1 Homo sapiens 117-121 17095152-11 2007 Based on these results, we conclude that HO-1 plays a major role in mediating cytoprotection and iron homeostasis against NO toxicity in immortalized and malignant oral keratinocytes. Iron 97-101 heme oxygenase 1 Homo sapiens 41-45 17339115-3 2007 The imidazole-dioxolanes were all selective for the HO-1 isozyme (inducible) and exhibited substantially lower activity toward the HO-2 isozyme (constitutive). imidazole-dioxolanes 4-24 heme oxygenase 1 Homo sapiens 52-56 17339115-4 2007 The corresponding imidazole-ketones and imidazole-alcohols showed selectivity toward HO-1 to a lesser degree than the similarly substituted imidazole-dioxolanes. imidazole-ketones 18-35 heme oxygenase 1 Homo sapiens 85-89 17339115-4 2007 The corresponding imidazole-ketones and imidazole-alcohols showed selectivity toward HO-1 to a lesser degree than the similarly substituted imidazole-dioxolanes. imidazole-alcohols 40-58 heme oxygenase 1 Homo sapiens 85-89 17339115-4 2007 The corresponding imidazole-ketones and imidazole-alcohols showed selectivity toward HO-1 to a lesser degree than the similarly substituted imidazole-dioxolanes. imidazole-dioxolanes 140-160 heme oxygenase 1 Homo sapiens 85-89 17275847-0 2007 Inhibition of heme oxygenase-1 protects against tissue injury in carbon tetrachloride exposed livers. Carbon Tetrachloride 65-85 heme oxygenase 1 Homo sapiens 14-30 17275847-2 2007 Heme oxygenase (HO-1) is an enzyme that is induced by heme as well as oxidative stress and has been reported to be involved in mediating protection against toxic liver injury. Heme 54-58 heme oxygenase 1 Homo sapiens 16-20 17275847-3 2007 The purpose of the present study was to specify the role of HO-1 in CCl(4)-hepatotoxicity. Carbon Tetrachloride 68-74 heme oxygenase 1 Homo sapiens 60-64 17275847-4 2007 METHODS AND RESULTS: We could demonstrate an up-regulation of HO-1 protein in CCl(4)-exposed liver tissue that reaches its maximum after 6 to 12 h, along with intrahepatic leukocyte accumulation and tissue injury. Cefaclor 78-81 heme oxygenase 1 Homo sapiens 62-66 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. Cefaclor 77-80 heme oxygenase 1 Homo sapiens 60-64 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. Cefaclor 77-80 heme oxygenase 1 Homo sapiens 200-204 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. Cefaclor 185-188 heme oxygenase 1 Homo sapiens 60-64 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. Cefaclor 185-188 heme oxygenase 1 Homo sapiens 200-204 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. tin protoporphyrin IX 219-240 heme oxygenase 1 Homo sapiens 60-64 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. tin protoporphyrin IX 219-240 heme oxygenase 1 Homo sapiens 200-204 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. tin protoporphyrin IX 242-249 heme oxygenase 1 Homo sapiens 60-64 17275847-5 2007 When animals were pretreated with hemin for augmentation of HO-1 expression, CCl(4)-exposure was associated with a reduction of intrahepatic leukocyte accumulation, while inhibition of CCl(4)-induced HO-1 expression by tin protoporphyrin-IX (SnPP-IX) enhanced leukocytic response. tin protoporphyrin IX 242-249 heme oxygenase 1 Homo sapiens 200-204 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Cefaclor 51-54 heme oxygenase 1 Homo sapiens 43-47 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Cefaclor 201-204 heme oxygenase 1 Homo sapiens 43-47 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Cefaclor 201-204 heme oxygenase 1 Homo sapiens 107-111 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Cefaclor 201-204 heme oxygenase 1 Homo sapiens 107-111 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Heme 253-257 heme oxygenase 1 Homo sapiens 43-47 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Heme 253-257 heme oxygenase 1 Homo sapiens 107-111 17275847-8 2007 CONCLUSIONS: Taken together, inhibition of HO-1 in CCl(4)-hepatotoxicity protected the liver, while higher HO-1 activity harmed liver tissue, most probably due to interference of the HO-1 pathway with CCl(4)-dependent metabolism via cytochrome P450 and heme overload-associated toxicity. Heme 253-257 heme oxygenase 1 Homo sapiens 107-111 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 30-33 heme oxygenase 1 Homo sapiens 92-108 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 30-33 heme oxygenase 1 Homo sapiens 110-114 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 30-33 heme oxygenase 1 Homo sapiens 268-272 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. gme 43-46 heme oxygenase 1 Homo sapiens 92-108 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. gme 43-46 heme oxygenase 1 Homo sapiens 110-114 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. gme 43-46 heme oxygenase 1 Homo sapiens 268-272 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Dopamine 56-58 heme oxygenase 1 Homo sapiens 268-272 17464197-4 2007 The inhibition was due to the induction of heme oxygenase (HO)-1 in LPS-activated macrophages, since blocking HO-1 activity with ZnPP, an HO-1 specific inhibitor, abolished hydroquinone"s NO inhibitory activity. zinc protoporphyrin 129-133 heme oxygenase 1 Homo sapiens 43-64 17466351-9 2007 Both 3,6-BPQ and 1,6-BPQ induced oxidative stress-associated genes (HMOX1, GCLC, GCLM, and SLC7A11), phase 2 enzyme genes (NQO1, NQO2, ALDH3A1), PAH metabolizing genes (CYP1B1, EPHX1, AKR1C1), and certain EGF receptor-associated genes (EGFR, IER3, ING1, SQSTM1 and TRIM16). benzo(a)pyrene-3,6-quinone 5-12 heme oxygenase 1 Homo sapiens 68-73 17466351-9 2007 Both 3,6-BPQ and 1,6-BPQ induced oxidative stress-associated genes (HMOX1, GCLC, GCLM, and SLC7A11), phase 2 enzyme genes (NQO1, NQO2, ALDH3A1), PAH metabolizing genes (CYP1B1, EPHX1, AKR1C1), and certain EGF receptor-associated genes (EGFR, IER3, ING1, SQSTM1 and TRIM16). 1,6-bpq 17-24 heme oxygenase 1 Homo sapiens 68-73 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 17-28 heme oxygenase 1 Homo sapiens 92-108 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 17-28 heme oxygenase 1 Homo sapiens 110-114 17400258-7 2007 In addition, the glutathione (GSH) analog (GME), blocks DA-induced superoxide accumulation, heme-oxygenase-1 (HO-1) expression and caspase-3 activation, and reduces cell death, while the glutathione synthetase inhibitor, buthionine sulfoximine, potentiates DA-induced HO-1 expression and cell death. Glutathione 17-28 heme oxygenase 1 Homo sapiens 268-272 17464175-1 2007 Curcumin is a polyphenolic compound possessing interesting anti-inflammatory and antioxidant properties and has the ability to induce the defensive protein heme oxygenase-1 (HO-1). Curcumin 0-8 heme oxygenase 1 Homo sapiens 156-172 17464175-1 2007 Curcumin is a polyphenolic compound possessing interesting anti-inflammatory and antioxidant properties and has the ability to induce the defensive protein heme oxygenase-1 (HO-1). Curcumin 0-8 heme oxygenase 1 Homo sapiens 174-178 17464175-1 2007 Curcumin is a polyphenolic compound possessing interesting anti-inflammatory and antioxidant properties and has the ability to induce the defensive protein heme oxygenase-1 (HO-1). polyphenolic compound 14-35 heme oxygenase 1 Homo sapiens 156-172 17464175-1 2007 Curcumin is a polyphenolic compound possessing interesting anti-inflammatory and antioxidant properties and has the ability to induce the defensive protein heme oxygenase-1 (HO-1). polyphenolic compound 14-35 heme oxygenase 1 Homo sapiens 174-178 17464175-5 2007 In additional experiments, an inhibitor of heme oxygenase activity (tin protoporphyrin IX, 10 microM) or siRNA for HO-1 were used to investigate the participation of HO-1 as a mediator of curcumin-induced effects. Curcumin 188-196 heme oxygenase 1 Homo sapiens 166-170 17464175-6 2007 Treatment with curcumin produced a marked induction of cardiac HO-1 in normothermic condition but cells were less responsive to the polyphenolic compound at low temperature. Curcumin 15-23 heme oxygenase 1 Homo sapiens 63-67 17464175-8 2007 Thus, curcumin added to Celsior preservation solution effectively prevents the damage caused by cold-storage; this effect involves the protective enzyme HO-1 but also other not yet identified mechanisms. Curcumin 6-14 heme oxygenase 1 Homo sapiens 153-157 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 18-22 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Carbon Monoxide 166-168 heme oxygenase 1 Homo sapiens 0-16 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Carbon Monoxide 166-168 heme oxygenase 1 Homo sapiens 18-22 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Bilirubin 174-183 heme oxygenase 1 Homo sapiens 0-16 17416426-2 2007 Heme oxygenase-1 (HO-1) is a key enzyme that is integral to the temporal and spatial regulation of the host response and, together with its products carbon monoxide (CO) and bilirubin, is crucial for maintaining homeostasis and the preservation of function and life. Bilirubin 174-183 heme oxygenase 1 Homo sapiens 18-22 17079780-4 2007 Heme oxygenase-1 expression increased dramatically in cytosolic and mitochondrial fractions of human alveolar (A549), or bronchial epithelial cells (Beas-2b) exposed to either hemin, lipopolysaccharide, or CSE. Hemin 176-181 heme oxygenase 1 Homo sapiens 0-16 17307160-4 2007 Treatment of A549 cells with LY 294002 (2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one, a PI3K inhibitor), an Akt inhibitor, and the dominant negative mutant of Akt (Akt DN) inhibited TGF-beta1-induced HO-1 expression and HO-1-luciferase activity. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 40-88 heme oxygenase 1 Homo sapiens 204-208 17079780-10 2007 Overexpression of heme oxygenase-1 inhibited CSE-induced Beas-2b cell death and preserved cellular ATP levels. Adenosine Triphosphate 99-102 heme oxygenase 1 Homo sapiens 18-34 17391133-2 2007 A functional dinucleotide repeat (GT)(n) polymorphism, within the HO-1 promoter, regulates gene expression; a short number of repeats (S-allele <25) increases transcription. Dinucleoside Phosphates 13-25 heme oxygenase 1 Homo sapiens 66-70 17392485-0 2007 Reduction of bilirubin by targeting human heme oxygenase-1 through siRNA. Bilirubin 13-22 heme oxygenase 1 Homo sapiens 42-58 17219054-0 2007 Induction of heme-oxygenase 1 requires the p38MAPK and PI3K pathways and suppresses apoptotic cell death following hypericin-mediated photodynamic therapy. hypericin 115-124 heme oxygenase 1 Homo sapiens 13-29 17219054-6 2007 Conversely, increasing HO-1 protein level by hemin prior to irradiation is cytoprotective. Hemin 45-50 heme oxygenase 1 Homo sapiens 23-27 17219054-8 2007 Altogether these results indicate that stimulation of HO-1 expression by hypericin-PDT is a cytoprotective mechanism governed by the p38(MAPK) and PI3K pathways, likely through the control of the nuclear availability of the Nrf2 pool. hypericin 73-82 heme oxygenase 1 Homo sapiens 54-58 17450221-7 2007 Additionally, the water-soluble fractions of UFPs were sufficient to induce the expression of F3, F2RL2, and heme oxygenase 1 (HMOX1). Water 18-23 heme oxygenase 1 Homo sapiens 109-125 17450221-7 2007 Additionally, the water-soluble fractions of UFPs were sufficient to induce the expression of F3, F2RL2, and heme oxygenase 1 (HMOX1). Water 18-23 heme oxygenase 1 Homo sapiens 127-132 17392485-3 2007 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme that generates bilirubin. Bilirubin 65-74 heme oxygenase 1 Homo sapiens 0-16 17392485-3 2007 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme that generates bilirubin. Bilirubin 65-74 heme oxygenase 1 Homo sapiens 18-22 17244614-0 2007 Cadmium-responsive element of the human heme oxygenase-1 gene mediates heat shock factor 1-dependent transcriptional activation. Cadmium 0-7 heme oxygenase 1 Homo sapiens 40-56 17235328-0 2007 Dimethylfumarate induces immunosuppression via glutathione depletion and subsequent induction of heme oxygenase 1. Dimethyl Fumarate 0-16 heme oxygenase 1 Homo sapiens 97-113 17235328-7 2007 Supplementation with exogenous glutathione (GSH), which is known to bind DMF, prevented both HO-1 induction as well as the anti-inflammatory effects of DMF. Glutathione 31-42 heme oxygenase 1 Homo sapiens 93-97 17235328-7 2007 Supplementation with exogenous glutathione (GSH), which is known to bind DMF, prevented both HO-1 induction as well as the anti-inflammatory effects of DMF. Glutathione 44-47 heme oxygenase 1 Homo sapiens 93-97 17235328-7 2007 Supplementation with exogenous glutathione (GSH), which is known to bind DMF, prevented both HO-1 induction as well as the anti-inflammatory effects of DMF. Dimethyl Fumarate 73-76 heme oxygenase 1 Homo sapiens 93-97 17235328-9 2007 These results suggest that DMF acts as active compound within the FAE mixture and at least partially mediates its immunomodulatory activity by the induction of the anti-inflammatory stress protein HO-1 ascribed to the functional depletion of reduced GSH. Dimethyl Fumarate 27-30 heme oxygenase 1 Homo sapiens 197-201 17235328-9 2007 These results suggest that DMF acts as active compound within the FAE mixture and at least partially mediates its immunomodulatory activity by the induction of the anti-inflammatory stress protein HO-1 ascribed to the functional depletion of reduced GSH. Glutathione 250-253 heme oxygenase 1 Homo sapiens 197-201 17408452-8 2007 HO (inducible HO-1, constitutive HO-2 and HO-3) is the rate-limiting enzyme in haeme catabolism, which leads to the generation of biliverdin, iron, and carbon monoxide. Biliverdine 130-140 heme oxygenase 1 Homo sapiens 14-18 17408452-8 2007 HO (inducible HO-1, constitutive HO-2 and HO-3) is the rate-limiting enzyme in haeme catabolism, which leads to the generation of biliverdin, iron, and carbon monoxide. Iron 142-146 heme oxygenase 1 Homo sapiens 14-18 17408452-8 2007 HO (inducible HO-1, constitutive HO-2 and HO-3) is the rate-limiting enzyme in haeme catabolism, which leads to the generation of biliverdin, iron, and carbon monoxide. Carbon Monoxide 152-167 heme oxygenase 1 Homo sapiens 14-18 17244614-2 2007 From a comparison of heavy metal responses of several human genes, it was noted that the heme oxygenase-1 (HO-1) gene is quite similar in the spectrum of metal response and induction kinetics to the heat shock protein 70 (HSP70) gene, suggesting a common regulatory mechanism shared by these genes. Metals 27-32 heme oxygenase 1 Homo sapiens 89-105 17244614-2 2007 From a comparison of heavy metal responses of several human genes, it was noted that the heme oxygenase-1 (HO-1) gene is quite similar in the spectrum of metal response and induction kinetics to the heat shock protein 70 (HSP70) gene, suggesting a common regulatory mechanism shared by these genes. Metals 27-32 heme oxygenase 1 Homo sapiens 107-111 17244614-2 2007 From a comparison of heavy metal responses of several human genes, it was noted that the heme oxygenase-1 (HO-1) gene is quite similar in the spectrum of metal response and induction kinetics to the heat shock protein 70 (HSP70) gene, suggesting a common regulatory mechanism shared by these genes. Metals 154-159 heme oxygenase 1 Homo sapiens 89-105 17244614-2 2007 From a comparison of heavy metal responses of several human genes, it was noted that the heme oxygenase-1 (HO-1) gene is quite similar in the spectrum of metal response and induction kinetics to the heat shock protein 70 (HSP70) gene, suggesting a common regulatory mechanism shared by these genes. Metals 154-159 heme oxygenase 1 Homo sapiens 107-111 17244614-3 2007 The cadmium-responsive element (CdRE) known to be responsible for the metal regulation of ho-1 formed complexes with proteins from heavy metal-treated HeLa cells in an electrophoretic mobility shift assay (EMSA). Cadmium 4-11 heme oxygenase 1 Homo sapiens 90-94 17244614-3 2007 The cadmium-responsive element (CdRE) known to be responsible for the metal regulation of ho-1 formed complexes with proteins from heavy metal-treated HeLa cells in an electrophoretic mobility shift assay (EMSA). cdre 32-36 heme oxygenase 1 Homo sapiens 90-94 17244614-3 2007 The cadmium-responsive element (CdRE) known to be responsible for the metal regulation of ho-1 formed complexes with proteins from heavy metal-treated HeLa cells in an electrophoretic mobility shift assay (EMSA). Metals 70-75 heme oxygenase 1 Homo sapiens 90-94 17244614-3 2007 The cadmium-responsive element (CdRE) known to be responsible for the metal regulation of ho-1 formed complexes with proteins from heavy metal-treated HeLa cells in an electrophoretic mobility shift assay (EMSA). Metals 137-142 heme oxygenase 1 Homo sapiens 90-94 17320768-3 2007 In the current study, we demonstrated that transient transfection of rat astroglia with human (h)ho-1 cDNA for 3 days significantly decreased intracellular cholesterol concentrations and increased levels of four oxysterol species (measured by GC/MS) compared to untreated control cultures and HO-1-transfected cells exposed to the HO inhibitor, tin mesoporphyrin (SnMP). Cholesterol 156-167 heme oxygenase 1 Homo sapiens 97-101 17257585-1 2007 Tin mesoporphyrin (SnMP), a competitive heme oxygenase (HO) inhibitor, also induces HO-1 mRNA and protein expression by a mechanism that is not fully understood. tin mesoporphyrin 0-17 heme oxygenase 1 Homo sapiens 84-88 17257585-1 2007 Tin mesoporphyrin (SnMP), a competitive heme oxygenase (HO) inhibitor, also induces HO-1 mRNA and protein expression by a mechanism that is not fully understood. tin mesoporphyrin 19-23 heme oxygenase 1 Homo sapiens 84-88 17257585-5 2007 Transfection of Bach1 shRNA in Hepa cells raised basal HO-1 expression significantly, and SnMP treatment further increased HO-1 mRNA. tin mesoporphyrin 90-94 heme oxygenase 1 Homo sapiens 123-127 17257585-6 2007 In conclusion, SnMP induces HO-1 expression not only by de-repressing the HO-1 promoter by binding Bach1, but also by accelerating Bach1 degradation. tin mesoporphyrin 15-19 heme oxygenase 1 Homo sapiens 28-32 17257585-6 2007 In conclusion, SnMP induces HO-1 expression not only by de-repressing the HO-1 promoter by binding Bach1, but also by accelerating Bach1 degradation. tin mesoporphyrin 15-19 heme oxygenase 1 Homo sapiens 74-78 17339489-8 2007 The same low H(2)O(2) concentration also induced the anti-inflammatory gene coding for heme oxygenase-1 (HO-1) and IL-6. Hydrogen Peroxide 13-21 heme oxygenase 1 Homo sapiens 87-103 17339489-8 2007 The same low H(2)O(2) concentration also induced the anti-inflammatory gene coding for heme oxygenase-1 (HO-1) and IL-6. Hydrogen Peroxide 13-21 heme oxygenase 1 Homo sapiens 105-109 17320768-3 2007 In the current study, we demonstrated that transient transfection of rat astroglia with human (h)ho-1 cDNA for 3 days significantly decreased intracellular cholesterol concentrations and increased levels of four oxysterol species (measured by GC/MS) compared to untreated control cultures and HO-1-transfected cells exposed to the HO inhibitor, tin mesoporphyrin (SnMP). Oxysterols 212-221 heme oxygenase 1 Homo sapiens 97-101 17320768-3 2007 In the current study, we demonstrated that transient transfection of rat astroglia with human (h)ho-1 cDNA for 3 days significantly decreased intracellular cholesterol concentrations and increased levels of four oxysterol species (measured by GC/MS) compared to untreated control cultures and HO-1-transfected cells exposed to the HO inhibitor, tin mesoporphyrin (SnMP). tin mesoporphyrin 345-362 heme oxygenase 1 Homo sapiens 97-101 17320768-3 2007 In the current study, we demonstrated that transient transfection of rat astroglia with human (h)ho-1 cDNA for 3 days significantly decreased intracellular cholesterol concentrations and increased levels of four oxysterol species (measured by GC/MS) compared to untreated control cultures and HO-1-transfected cells exposed to the HO inhibitor, tin mesoporphyrin (SnMP). tin mesoporphyrin 364-368 heme oxygenase 1 Homo sapiens 97-101 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). linoleoyl tyrosine 253-271 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). Leu-Thr 273-275 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). linoleoyl tyrosine cholesterol ester 278-314 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). ltc 316-319 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). linoleoyl tyrosine 278-296 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). deoxyguanosyl ester 344-363 heme oxygenase 1 Homo sapiens 152-156 17320768-4 2007 Relative to control preparations, oxidative stress was augmented in mitochondria (isolated by subcellular fractionation) and culture media derived from HO-1-transfected astrocytes, as evidenced by enhanced oxidation of the synthetic reporter molecules, linoleoyl tyrosine (LT), linoleoyl tyrosine cholesterol ester (LTC), or linoleoyl tyrosine deoxyguanosyl ester (LTG; measured by GC/MS and LC/MS/MS). Lamotrigine 365-368 heme oxygenase 1 Homo sapiens 152-156 17320768-6 2007 In AD and other pathological states, glial HO-1 induction may transduce ambient noxious stimuli (e.g., beta-amyloid) into altered patterns of glial sterol metabolism which, in turn, may affect neuronal membrane turnover, survival, and adaptability. Sterols 148-154 heme oxygenase 1 Homo sapiens 43-47 17204476-1 2007 Heme oxygenase-1 is a highly inducible gene, the product of which catalyzes breakdown of the prooxidant heme. Heme 104-108 heme oxygenase 1 Homo sapiens 0-16 17204476-5 2007 Two mutations within HS-2 combined with a third mutation of the proximal E box abolished hemin- and cadmium-driven heme oxygenase-1 promoter activation, suggesting that these three sites synergized for maximal heme oxygenase-1 induction. Hemin 89-94 heme oxygenase 1 Homo sapiens 115-131 17204476-5 2007 Two mutations within HS-2 combined with a third mutation of the proximal E box abolished hemin- and cadmium-driven heme oxygenase-1 promoter activation, suggesting that these three sites synergized for maximal heme oxygenase-1 induction. Hemin 89-94 heme oxygenase 1 Homo sapiens 210-226 17204476-5 2007 Two mutations within HS-2 combined with a third mutation of the proximal E box abolished hemin- and cadmium-driven heme oxygenase-1 promoter activation, suggesting that these three sites synergized for maximal heme oxygenase-1 induction. Cadmium 100-107 heme oxygenase 1 Homo sapiens 115-131 17204476-5 2007 Two mutations within HS-2 combined with a third mutation of the proximal E box abolished hemin- and cadmium-driven heme oxygenase-1 promoter activation, suggesting that these three sites synergized for maximal heme oxygenase-1 induction. Cadmium 100-107 heme oxygenase 1 Homo sapiens 210-226 17160582-1 2007 Recombinant truncated human heme oxygenase-1 (hHO-1) expressed in Escherichia coli was efficiently separated and purified from feedstock by DEAE-ion exchange expanded bed adsorption. 2-diethylaminoethanol 140-144 heme oxygenase 1 Homo sapiens 28-44 17160582-1 2007 Recombinant truncated human heme oxygenase-1 (hHO-1) expressed in Escherichia coli was efficiently separated and purified from feedstock by DEAE-ion exchange expanded bed adsorption. 2-diethylaminoethanol 140-144 heme oxygenase 1 Homo sapiens 46-51 17279724-2 2007 It is known to inhibit heme-oxygenase-1 (HO-1), resulting in suppressed biliverdin/bilirubin production accompanying lowered antioxidative capacity. Biliverdine 72-82 heme oxygenase 1 Homo sapiens 23-39 17279724-2 2007 It is known to inhibit heme-oxygenase-1 (HO-1), resulting in suppressed biliverdin/bilirubin production accompanying lowered antioxidative capacity. Bilirubin 83-92 heme oxygenase 1 Homo sapiens 23-39 17160582-2 2007 Protocol optimization of hHO-1 on DEAE adsorbent resulted in adsorption in 0 M NaCl and elution in 150 mM NaCl at a pH of 8.5. 2-diethylaminoethanol 34-38 heme oxygenase 1 Homo sapiens 25-30 17160582-2 2007 Protocol optimization of hHO-1 on DEAE adsorbent resulted in adsorption in 0 M NaCl and elution in 150 mM NaCl at a pH of 8.5. Sodium Chloride 79-83 heme oxygenase 1 Homo sapiens 25-30 17160582-2 2007 Protocol optimization of hHO-1 on DEAE adsorbent resulted in adsorption in 0 M NaCl and elution in 150 mM NaCl at a pH of 8.5. Sodium Chloride 106-110 heme oxygenase 1 Homo sapiens 25-30 16924499-9 2007 After the initial increase in intracellular peroxides, the heat shock protein and antioxidant heme oxygenase-1 (HO-1) was upregulated. Peroxides 44-53 heme oxygenase 1 Homo sapiens 94-110 16924499-9 2007 After the initial increase in intracellular peroxides, the heat shock protein and antioxidant heme oxygenase-1 (HO-1) was upregulated. Peroxides 44-53 heme oxygenase 1 Homo sapiens 112-116 16924499-11 2007 NAC attenuated HO-1 upregulation, confirming the time course analysis. Acetylcysteine 0-3 heme oxygenase 1 Homo sapiens 15-19 17291539-0 2007 Preconditioning with hyperbaric oxygen induces tolerance against oxidative injury via increased expression of heme oxygenase-1 in primary cultured spinal cord neurons. Oxygen 32-38 heme oxygenase 1 Homo sapiens 110-126 17225922-9 2007 Micelle formulations prepared with ethanol/Tween40 resulted in the lowest LDH release, the highest carotenoid uptake and the lowest stress response (changes in HO-1 mRNA expression). Ethanol 35-42 heme oxygenase 1 Homo sapiens 160-164 17225922-9 2007 Micelle formulations prepared with ethanol/Tween40 resulted in the lowest LDH release, the highest carotenoid uptake and the lowest stress response (changes in HO-1 mRNA expression). polysorbate 40 43-50 heme oxygenase 1 Homo sapiens 160-164 17325212-3 2007 The boy was a homozygous carrier of short alleles of the heme oxygenase-1 (HO-1) gene GT dinucleotide-repeat promoter polymorphism, which is associated with increased activity and inducibility of the heme-degrading enzyme HO-1, which catalyzes the production of bilirubin. Heme 57-61 heme oxygenase 1 Homo sapiens 75-79 17325212-3 2007 The boy was a homozygous carrier of short alleles of the heme oxygenase-1 (HO-1) gene GT dinucleotide-repeat promoter polymorphism, which is associated with increased activity and inducibility of the heme-degrading enzyme HO-1, which catalyzes the production of bilirubin. Heme 57-61 heme oxygenase 1 Homo sapiens 222-226 17325212-3 2007 The boy was a homozygous carrier of short alleles of the heme oxygenase-1 (HO-1) gene GT dinucleotide-repeat promoter polymorphism, which is associated with increased activity and inducibility of the heme-degrading enzyme HO-1, which catalyzes the production of bilirubin. Bilirubin 262-271 heme oxygenase 1 Homo sapiens 57-73 17325212-3 2007 The boy was a homozygous carrier of short alleles of the heme oxygenase-1 (HO-1) gene GT dinucleotide-repeat promoter polymorphism, which is associated with increased activity and inducibility of the heme-degrading enzyme HO-1, which catalyzes the production of bilirubin. Bilirubin 262-271 heme oxygenase 1 Homo sapiens 75-79 17325212-5 2007 Because bilirubin production plays a critical role during the neonatal period, the HO-1 promoter polymorphism may be an important genetic factor for the clinical outcome of neonatal hyperbilirubinemia. Bilirubin 8-17 heme oxygenase 1 Homo sapiens 83-87 17291539-7 2007 Treatment with tin-mesoporphyrin IX (SnMP), a specific HO-1 inhibitor, before HBO pretreatment abolished the HBO-induced adaptive protection noted in the cultured spinal neurons. tin-mesoporphyrin ix 15-35 heme oxygenase 1 Homo sapiens 55-59 17291539-7 2007 Treatment with tin-mesoporphyrin IX (SnMP), a specific HO-1 inhibitor, before HBO pretreatment abolished the HBO-induced adaptive protection noted in the cultured spinal neurons. tin mesoporphyrin 37-41 heme oxygenase 1 Homo sapiens 55-59 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Heme 54-58 heme oxygenase 1 Homo sapiens 72-76 16517059-0 2007 Ascorbic acid differentially modulates the induction of heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and glutathione S-transferase Ya by As(3+), Cd(2+) and Cr(6+). Ascorbic Acid 0-13 heme oxygenase 1 Homo sapiens 56-72 16517059-0 2007 Ascorbic acid differentially modulates the induction of heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and glutathione S-transferase Ya by As(3+), Cd(2+) and Cr(6+). Cadmium 151-153 heme oxygenase 1 Homo sapiens 56-72 16517059-0 2007 Ascorbic acid differentially modulates the induction of heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1 and glutathione S-transferase Ya by As(3+), Cd(2+) and Cr(6+). Chromium 162-164 heme oxygenase 1 Homo sapiens 56-72 16517059-7 2007 Hence, it is tempting to speculate that AA may potentiate the therapeutic efficacy of As(3+) by enhancing the expression of HO-1, Nqo1, and Gst ya while acting as a potent antioxidant. Arsenic(3+) ion 86-92 heme oxygenase 1 Homo sapiens 124-128 16980551-2 2007 HO-1 catalyzes the degradation of heme, a potent oxidant, into biliverdin, iron, and carbon monoxide (CO). Biliverdine 63-73 heme oxygenase 1 Homo sapiens 0-4 16980551-2 2007 HO-1 catalyzes the degradation of heme, a potent oxidant, into biliverdin, iron, and carbon monoxide (CO). Iron 75-79 heme oxygenase 1 Homo sapiens 0-4 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Heme 134-138 heme oxygenase 1 Homo sapiens 54-70 16980551-2 2007 HO-1 catalyzes the degradation of heme, a potent oxidant, into biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 85-100 heme oxygenase 1 Homo sapiens 0-4 16980551-2 2007 HO-1 catalyzes the degradation of heme, a potent oxidant, into biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 102-104 heme oxygenase 1 Homo sapiens 0-4 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Heme 134-138 heme oxygenase 1 Homo sapiens 72-76 16980551-3 2007 These downstream products of heme catabolism have recently been found to mediate the antioxidant, antiapoptotic, antiproliferative, vasodilatory, and anti-inflammatory properties of HO-1. Heme 29-33 heme oxygenase 1 Homo sapiens 182-186 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Biliverdine 238-256 heme oxygenase 1 Homo sapiens 54-70 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Biliverdine 238-256 heme oxygenase 1 Homo sapiens 72-76 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Carbon Monoxide 258-273 heme oxygenase 1 Homo sapiens 54-70 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Carbon Monoxide 258-273 heme oxygenase 1 Homo sapiens 72-76 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Iron 279-283 heme oxygenase 1 Homo sapiens 54-70 16990612-3 2007 One protein providing such cytoprotective activity is heme oxygenase-1 (HO-1), an enzyme that catalyzes the rate-limiting reaction in heme catabolism (i.e., the oxidative cleavage of b-type heme molecules to yield equimolar quantities of biliverdin IXalpha, carbon monoxide, and iron). Iron 279-283 heme oxygenase 1 Homo sapiens 72-76 16838348-4 2007 The aim of this study was to investigate the effects of zinc oxide-eugenol-based root canal sealer N2 and epoxy resin-based root canal sealer Topseal on the expression of HO-1 protein in cultured human gingival fibroblasts (HGFs). Zinc Oxide 56-66 heme oxygenase 1 Homo sapiens 171-175 17018862-7 2007 Because CML cells are known to produce high levels of intracellular reactive oxygen species, overexpression of heme oxygenase-1 resulting from inhibition of Bach2 activity may contribute to their genomic instability and leukemic phenotype. Reactive Oxygen Species 68-91 heme oxygenase 1 Homo sapiens 111-127 17258074-9 2007 CONCLUSIONS: Arsenic trioxide-induced renal toxicity is strongly associated with the increased expression of HMOX1, and the cytotoxic mechanisms of arsenic trioxide involves reactive oxygen species production as well as another pathway. Arsenic Trioxide 13-29 heme oxygenase 1 Homo sapiens 109-114 17169865-6 2007 Interestingly, an increased expression of HO-1, a redox-sensitive stress protein, was observed in NO2-exposed NHBE cells at 24 h. Since neutrophils (PMNs) play an active role in acute lung inflammation and resultant oxidative injury, we also investigated changes in human PMN-NHBE cell interactions. Nitrogen Dioxide 98-101 heme oxygenase 1 Homo sapiens 42-46 17229906-4 2007 Finally, current views regarding the molecular basis for heme-induced upregulation of HO-1 are discussed. Heme 57-61 heme oxygenase 1 Homo sapiens 86-90 16838348-4 2007 The aim of this study was to investigate the effects of zinc oxide-eugenol-based root canal sealer N2 and epoxy resin-based root canal sealer Topseal on the expression of HO-1 protein in cultured human gingival fibroblasts (HGFs). Eugenol 67-74 heme oxygenase 1 Homo sapiens 171-175 16838348-8 2007 However, BSO enhanced the N2-induced HO-1 protein level by up to twofold (p < 0.05). Nitrogen 26-28 heme oxygenase 1 Homo sapiens 37-41 17277740-15 2007 Both vitreous and TGF-beta signals increased HO-1 expression via ROS but the latter were not involved in vitreous-mediated MT expression. Reactive Oxygen Species 65-68 heme oxygenase 1 Homo sapiens 45-49 17277740-0 2007 Vitreous induces heme oxygenase-1 expression mediated by transforming growth factor-beta and reactive oxygen species generation in human retinal pigment epithelial cells. Reactive Oxygen Species 93-116 heme oxygenase 1 Homo sapiens 17-33 17184579-5 2007 The expression of HO-1 mRNA in PASMC was analyzed by fluorescent real-time quantitative PCR, and the proliferating cell nuclear antigen and caspase-3 were examined by immunocytochemical analysis. pasmc 31-36 heme oxygenase 1 Homo sapiens 18-22 17277740-10 2007 Addition of inhibitors of TGF-beta signaling (SB431542 or TGF-beta-neutralizing antibodies) decreased the vitreous induction of HO-1. 4-(5-benzo(1,3)dioxol-5-yl-4-pyridin-2-yl-1H-imidazol-2-yl)benzamide 46-54 heme oxygenase 1 Homo sapiens 128-132 17277740-11 2007 Several reactive oxygen species (ROS) quenchers inhibited the TGF-beta-induced or vitreous-induced elevation of HO-1 mRNA but had no effect on vitreous-mediated induction of MT expression. Reactive Oxygen Species 8-31 heme oxygenase 1 Homo sapiens 112-116 17277740-11 2007 Several reactive oxygen species (ROS) quenchers inhibited the TGF-beta-induced or vitreous-induced elevation of HO-1 mRNA but had no effect on vitreous-mediated induction of MT expression. Reactive Oxygen Species 33-36 heme oxygenase 1 Homo sapiens 112-116 17277740-12 2007 Inhibitors of the mitogen-activated protein kinase (p38MAPK; SB203580) and Jun N-terminal kinase (JNK; SP600125) pathways inhibited vitreous-induction of HO-1. SB 203580 61-69 heme oxygenase 1 Homo sapiens 154-158 17277740-12 2007 Inhibitors of the mitogen-activated protein kinase (p38MAPK; SB203580) and Jun N-terminal kinase (JNK; SP600125) pathways inhibited vitreous-induction of HO-1. pyrazolanthrone 103-111 heme oxygenase 1 Homo sapiens 154-158 17189831-6 2007 Incubation with the thiol antioxidant N-acetylcysteine strongly inhibited both the Nrf2 accumulation and the expression of Nrf2-regulated genes such as HO-1, GCLM, and SQSTM1. Sulfhydryl Compounds 20-25 heme oxygenase 1 Homo sapiens 152-156 17189831-6 2007 Incubation with the thiol antioxidant N-acetylcysteine strongly inhibited both the Nrf2 accumulation and the expression of Nrf2-regulated genes such as HO-1, GCLM, and SQSTM1. Acetylcysteine 38-54 heme oxygenase 1 Homo sapiens 152-156 17569214-1 2007 Curcumin possesses anti-inflammatory activity and is a potent inhibitor of reactive-oxygen-generating enzymes such as lipoxygenase/cyclooxygenase, xanthine dehydrogenase/oxidase, and inducible nitric oxide synthase (iNOS); it is an effective inducer of heme oxygenase-1. Curcumin 0-8 heme oxygenase 1 Homo sapiens 253-269 17196236-0 2007 Contributions of reactive oxygen species and mitogen-activated protein kinase signaling in arsenite-stimulated hemeoxygenase-1 production. Reactive Oxygen Species 17-40 heme oxygenase 1 Homo sapiens 111-126 17184579-7 2007 Meanwhile, hypoxia induced HO-1 expression in PASMC and promoted CO production from PASMC, which inhibited PASMC proliferation and regulated PASMC apoptosis. pasmc 46-51 heme oxygenase 1 Homo sapiens 27-31 17184579-8 2007 NO upregulated the expression of HO-1 mRNA in hypoxic PASMC; NO also inhibited proliferation and promoted apoptosis of hypoxic PASMC, possibly by regulating the production of CO. pasmc 54-59 heme oxygenase 1 Homo sapiens 33-37 17569214-1 2007 Curcumin possesses anti-inflammatory activity and is a potent inhibitor of reactive-oxygen-generating enzymes such as lipoxygenase/cyclooxygenase, xanthine dehydrogenase/oxidase, and inducible nitric oxide synthase (iNOS); it is an effective inducer of heme oxygenase-1. reactive-oxygen 75-90 heme oxygenase 1 Homo sapiens 253-269 17184579-8 2007 NO upregulated the expression of HO-1 mRNA in hypoxic PASMC; NO also inhibited proliferation and promoted apoptosis of hypoxic PASMC, possibly by regulating the production of CO. pasmc 127-132 heme oxygenase 1 Homo sapiens 33-37 17264037-10 2007 This oxygen species may induce redox-sensitive gene transcription such as HO-1. Oxygen 5-11 heme oxygenase 1 Homo sapiens 74-78 17265967-6 2007 Vanadium oxide leads to an induction of heme oxygenase 1 (HO-1) in a dose dependent manner in ECV304 cells whereas a reduction in protein levels can be observed for the epithelial cells (A549). vanadium pentoxide 0-14 heme oxygenase 1 Homo sapiens 40-56 17245120-5 2007 This combined with experimental data showing anti-atherosclerotic properties of the enzyme heme oxygenase-1 encouraged us to hypothesize that bilirubin and its precursor biliverdin, would act to ameliorate components of atherosclerosis, in a manner similar to what has been shown with HO-1. Bilirubin 142-151 heme oxygenase 1 Homo sapiens 91-107 17245120-5 2007 This combined with experimental data showing anti-atherosclerotic properties of the enzyme heme oxygenase-1 encouraged us to hypothesize that bilirubin and its precursor biliverdin, would act to ameliorate components of atherosclerosis, in a manner similar to what has been shown with HO-1. Biliverdine 170-180 heme oxygenase 1 Homo sapiens 91-107 17115887-9 2007 Among these, the heme oxygenase-1/carbon monoxide system has emerged as a major intracellular antiapoptotic mechanism. Carbon Monoxide 34-49 heme oxygenase 1 Homo sapiens 17-33 17095719-4 2007 Three genes involved in iron-heme homeostasis, CD163, HO-1, and transferrin receptor, were further analyzed in 40 independent plaques. Iron 24-28 heme oxygenase 1 Homo sapiens 54-84 17095719-6 2007 The expression of HO-1 and CD163 correlated with tissue iron content but iron itself was not associated with the symptom status. Iron 56-60 heme oxygenase 1 Homo sapiens 18-22 17265967-6 2007 Vanadium oxide leads to an induction of heme oxygenase 1 (HO-1) in a dose dependent manner in ECV304 cells whereas a reduction in protein levels can be observed for the epithelial cells (A549). vanadium pentoxide 0-14 heme oxygenase 1 Homo sapiens 58-62 17143561-0 2007 Curcumin induces heme oxygenase 1 through generation of reactive oxygen species, p38 activation and phosphatase inhibition. Curcumin 0-8 heme oxygenase 1 Homo sapiens 17-33 17141778-8 2007 The nitric oxide (NO) donor-SNP ameliorates LPS-induced ALI, which may be related to the induction of HO-1 and the subsequent inhibition of iNOS. Nitric Oxide 4-16 heme oxygenase 1 Homo sapiens 102-106 17143561-0 2007 Curcumin induces heme oxygenase 1 through generation of reactive oxygen species, p38 activation and phosphatase inhibition. Reactive Oxygen Species 56-79 heme oxygenase 1 Homo sapiens 17-33 17143561-1 2007 Curcumin is a naturally occurring compound which is known to induce heme oxygenase 1 (HO-1), although the underlying mechanism has not been fully elucidated. Curcumin 0-8 heme oxygenase 1 Homo sapiens 68-84 17143561-1 2007 Curcumin is a naturally occurring compound which is known to induce heme oxygenase 1 (HO-1), although the underlying mechanism has not been fully elucidated. Curcumin 0-8 heme oxygenase 1 Homo sapiens 86-90 17143561-2 2007 This study investigates in detail the mechanism of HO-1 induction by curcumin in human hepatoma cells. Curcumin 69-77 heme oxygenase 1 Homo sapiens 51-55 17143561-4 2007 Curcumin was found to induce HO-1 at doses of 10 to 25 microM. Curcumin 0-8 heme oxygenase 1 Homo sapiens 29-33 17143561-5 2007 At both non-toxic and toxic doses, HO-1 induction was found to correlate with production of reactive oxygen species (ROS), suggesting a causative relationship. Reactive Oxygen Species 92-115 heme oxygenase 1 Homo sapiens 35-39 17143561-5 2007 At both non-toxic and toxic doses, HO-1 induction was found to correlate with production of reactive oxygen species (ROS), suggesting a causative relationship. Reactive Oxygen Species 117-120 heme oxygenase 1 Homo sapiens 35-39 17143561-6 2007 This was reinforced by the finding that pretreatment with the antioxidants N-acetylcysteine, vitamin E and catalase prevented HO-1 induction by curcumin. Acetylcysteine 75-91 heme oxygenase 1 Homo sapiens 126-130 17143561-6 2007 This was reinforced by the finding that pretreatment with the antioxidants N-acetylcysteine, vitamin E and catalase prevented HO-1 induction by curcumin. Vitamin E 93-102 heme oxygenase 1 Homo sapiens 126-130 17143561-6 2007 This was reinforced by the finding that pretreatment with the antioxidants N-acetylcysteine, vitamin E and catalase prevented HO-1 induction by curcumin. Curcumin 144-152 heme oxygenase 1 Homo sapiens 126-130 17143561-10 2007 A panel of kinase inhibitors was used to examine the contribution of MAP kinases to the induction of HO-1 by curcumin. Curcumin 109-117 heme oxygenase 1 Homo sapiens 101-105 17143561-13 2007 In conclusion, curcumin treatment results in ROS generation, activation of Nrf2 and MAP kinases and the inhibition of phosphatase activity in hepatocytes, and when curcumin is not administered in toxic doses, these multiple pathways converge to induce HO-1. Curcumin 15-23 heme oxygenase 1 Homo sapiens 252-256 17191021-9 2007 Moreover, while hemoxygenase-1 (HO-1) expression was induced by resveratrol in human umbilical vein endothelial cells, neither treatment with the HO-1 inhibitor tin-protoporphyrin IX nor siRNA-directed knockdown of HO-1 had any effect on the inhibition of MCP-1 mRNA or protein secretion by resveratrol. Resveratrol 64-75 heme oxygenase 1 Homo sapiens 16-36 17825497-2 2007 One form of this of this enzyme, heme oxygenase-1, is inducible by numerous agents which promote oxidative stress, and is now known to provide important antioxidant protection, as demonstrated in many rodent models of free radical-mediated pathogenesis, and suggested by epidemiology observing favorable health outcomes in individuals carrying high-expression alleles of the HO-1 gene. Free Radicals 218-230 heme oxygenase 1 Homo sapiens 33-49 17018578-0 2007 Motexafin gadolinium-induced cell death correlates with heme oxygenase-1 expression and inhibition of P450 reductase-dependent activities. Motexafin 0-9 heme oxygenase 1 Homo sapiens 56-72 18274635-5 2007 The results show that production of both NO and HO-1 is fairly dose dependent but is particularly elevated using human plasma after transient exposure to a medium ozone concentration. Ozone 163-168 heme oxygenase 1 Homo sapiens 48-52 17018578-0 2007 Motexafin gadolinium-induced cell death correlates with heme oxygenase-1 expression and inhibition of P450 reductase-dependent activities. Gadolinium 10-20 heme oxygenase 1 Homo sapiens 56-72 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Heme 39-43 heme oxygenase 1 Homo sapiens 0-16 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Heme 39-43 heme oxygenase 1 Homo sapiens 18-21 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Biliverdine 47-57 heme oxygenase 1 Homo sapiens 0-16 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Biliverdine 47-57 heme oxygenase 1 Homo sapiens 18-21 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Carbon Monoxide 59-61 heme oxygenase 1 Homo sapiens 0-16 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Carbon Monoxide 59-61 heme oxygenase 1 Homo sapiens 18-21 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Iron 72-76 heme oxygenase 1 Homo sapiens 0-16 17018578-1 2007 Heme oxygenase-1 (HO1), which oxidizes heme to biliverdin, CO, and free iron, conveys protection against oxidative stress and is antiapoptotic. Iron 72-76 heme oxygenase 1 Homo sapiens 18-21 17018578-5 2007 MGd used in combination with tin protoporphyrin IX, an inhibitor of HO1, resulted in synergistic cell killing. tin protoporphyrin IX 29-50 heme oxygenase 1 Homo sapiens 68-71 17018578-7 2007 We demonstrate that inhibition of HO1 reflects an interaction of MGd with NADPH-cytochrome P450 reductase, the electron donor for HO1, that results in diversion of reducing equivalents from heme oxidation to oxygen reduction. Heme 190-194 heme oxygenase 1 Homo sapiens 34-37 17018578-7 2007 We demonstrate that inhibition of HO1 reflects an interaction of MGd with NADPH-cytochrome P450 reductase, the electron donor for HO1, that results in diversion of reducing equivalents from heme oxidation to oxygen reduction. Heme 190-194 heme oxygenase 1 Homo sapiens 130-133 17018578-7 2007 We demonstrate that inhibition of HO1 reflects an interaction of MGd with NADPH-cytochrome P450 reductase, the electron donor for HO1, that results in diversion of reducing equivalents from heme oxidation to oxygen reduction. Oxygen 208-214 heme oxygenase 1 Homo sapiens 34-37 17108707-6 2007 Interestingly, pre-induction of heme oxygenase (HO)-1 by hemin prevented dialysis membrane-induced monocyte apoptosis, whereas inhibition of HO-1 activity (treatment with tin protoporphyrin, SN-P) enhanced dialysis membrane-induced monocyte apoptosis. protoporphyrin IX 175-189 heme oxygenase 1 Homo sapiens 141-145 17018578-7 2007 We demonstrate that inhibition of HO1 reflects an interaction of MGd with NADPH-cytochrome P450 reductase, the electron donor for HO1, that results in diversion of reducing equivalents from heme oxidation to oxygen reduction. Oxygen 208-214 heme oxygenase 1 Homo sapiens 130-133 17942419-4 2007 HMOX1 induction, elicited by arsenite-mediated oxidative stress, follows inactivation of BACH1 and precedes activation of NRF2. arsenite 29-37 heme oxygenase 1 Homo sapiens 0-5 17122584-1 2006 BACKGROUND: The volatile anesthetic isoflurane induces hypoxia inducible factor (HIF)-1-responsive genes heme oxygenase 1, inducible nitric oxide synthase, and vascular endothelial growth factor (VEGF) expression. Isoflurane 36-46 heme oxygenase 1 Homo sapiens 105-154 17122584-10 2006 Isoflurane stimulated heme oxygenase 1, inducible nitric oxide synthase, and VEGF mRNA expression in a concentration-dependent manner, and inactivation of HIF-1alpha attenuated the induction of VEGF mRNA by isoflurane. Isoflurane 0-10 heme oxygenase 1 Homo sapiens 22-71 17122584-11 2006 CONCLUSION: Isoflurane can up-regulate HIF-1alpha and enhance HIF-1-responsive genes heme oxygenase 1, inducible nitric oxide synthase, and VEGF mRNA expression in Hep3B cells. Isoflurane 12-22 heme oxygenase 1 Homo sapiens 85-134 17156779-9 2006 Treatment with CdCl(2) (to induce haem oxygenase, HO-1) enhanced HCP1 expression and increased haem uptake into the cells. Cadmium Chloride 15-22 heme oxygenase 1 Homo sapiens 50-54 16881871-2 2006 We have previously shown that PDTC (pyrrolidine dithiocarbamate), an HO-1 (haem oxygenase-1) donor, improves intestinal microvascular perfusion. pyrrolidine dithiocarbamic acid 30-34 heme oxygenase 1 Homo sapiens 69-73 17384282-2 2006 15-Deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), an endogenous ligand of peroxisome proliferator-activated receptor (PPAR) gamma, has been reported to upregulate VEGF synthesis through the induction of heme oxygenase (HO)-1. 14-prostaglandin j2 17-36 heme oxygenase 1 Homo sapiens 202-223 17384282-2 2006 15-Deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), an endogenous ligand of peroxisome proliferator-activated receptor (PPAR) gamma, has been reported to upregulate VEGF synthesis through the induction of heme oxygenase (HO)-1. 15-deoxy-delta(12,14)-prostaglandin J2 38-46 heme oxygenase 1 Homo sapiens 202-223 17384282-3 2006 In this work, we found that treatment of human breast cancer (MCF-7) cells with 15d-PGJ2 led to time-dependent increases in the expression of HO-1. 15-deoxy-delta(12,14)-prostaglandin J2 80-88 heme oxygenase 1 Homo sapiens 142-146 17384282-6 2006 The induction of HO-1 expression preceded the upregulation of VEGF in MCF-7 cells stimulated with 15d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 98-106 heme oxygenase 1 Homo sapiens 17-21 17384282-8 2006 To determine whether the induction of HO-1 is responsible for ERK1/2 activation, the HO-1 inhibitor, zinc protoporphyrin (ZnPP) was used. zinc protoporphyrin 101-120 heme oxygenase 1 Homo sapiens 38-42 17384282-10 2006 These results suggest that 15d-PGJ2 upregulates VEGF expression via induction of HO-1 and ERK-1 and -2 phosphorylation, which may contribute to increased angiogenesis of the tumor cells. 15-deoxy-delta(12,14)-prostaglandin J2 27-35 heme oxygenase 1 Homo sapiens 81-85 16881871-2 2006 We have previously shown that PDTC (pyrrolidine dithiocarbamate), an HO-1 (haem oxygenase-1) donor, improves intestinal microvascular perfusion. pyrrolidine dithiocarbamic acid 30-34 heme oxygenase 1 Homo sapiens 75-91 16881871-2 2006 We have previously shown that PDTC (pyrrolidine dithiocarbamate), an HO-1 (haem oxygenase-1) donor, improves intestinal microvascular perfusion. pyrrolidine dithiocarbamic acid 36-63 heme oxygenase 1 Homo sapiens 69-73 16881871-2 2006 We have previously shown that PDTC (pyrrolidine dithiocarbamate), an HO-1 (haem oxygenase-1) donor, improves intestinal microvascular perfusion. pyrrolidine dithiocarbamic acid 36-63 heme oxygenase 1 Homo sapiens 75-91 17020887-0 2006 Resistance to nitric oxide-induced necrosis in heme oxygenase-1 overexpressing pulmonary epithelial cells associated with decreased lipid peroxidation. Nitric Oxide 14-26 heme oxygenase 1 Homo sapiens 47-63 17065227-0 2006 Role of Bach1 and Nrf2 in up-regulation of the heme oxygenase-1 gene by cobalt protoporphyrin. cobaltiprotoporphyrin 72-93 heme oxygenase 1 Homo sapiens 47-63 17168739-5 2006 Concurrently, the hypoxia-inducible factor (HIF) has also been shown in previous studies to regulate intracellular iron by binding to HIF-responsive elements (HREs) that are located within the genes of iron-related proteins such as TfR and heme oxygenase-1 (HO-1). Iron 115-119 heme oxygenase 1 Homo sapiens 240-256 17168739-5 2006 Concurrently, the hypoxia-inducible factor (HIF) has also been shown in previous studies to regulate intracellular iron by binding to HIF-responsive elements (HREs) that are located within the genes of iron-related proteins such as TfR and heme oxygenase-1 (HO-1). Iron 115-119 heme oxygenase 1 Homo sapiens 258-262 17168739-5 2006 Concurrently, the hypoxia-inducible factor (HIF) has also been shown in previous studies to regulate intracellular iron by binding to HIF-responsive elements (HREs) that are located within the genes of iron-related proteins such as TfR and heme oxygenase-1 (HO-1). Iron 202-206 heme oxygenase 1 Homo sapiens 240-256 17168739-5 2006 Concurrently, the hypoxia-inducible factor (HIF) has also been shown in previous studies to regulate intracellular iron by binding to HIF-responsive elements (HREs) that are located within the genes of iron-related proteins such as TfR and heme oxygenase-1 (HO-1). Iron 202-206 heme oxygenase 1 Homo sapiens 258-262 17020887-4 2006 Compared with the Neo control, the HO-1-overexpressing cells also showed significantly less lipid peroxide formation and decreased perturbation of transition metal oxidation and coordination states following a cytotoxic NO exposure. neo 18-21 heme oxygenase 1 Homo sapiens 35-39 17020887-4 2006 Compared with the Neo control, the HO-1-overexpressing cells also showed significantly less lipid peroxide formation and decreased perturbation of transition metal oxidation and coordination states following a cytotoxic NO exposure. Lipid Peroxides 92-106 heme oxygenase 1 Homo sapiens 35-39 17020887-4 2006 Compared with the Neo control, the HO-1-overexpressing cells also showed significantly less lipid peroxide formation and decreased perturbation of transition metal oxidation and coordination states following a cytotoxic NO exposure. Metals 158-163 heme oxygenase 1 Homo sapiens 35-39 17020887-5 2006 These effects were blocked by the HO-1 inhibitors Zn- and Sn-protoporphyrin IX. Zinc 50-52 heme oxygenase 1 Homo sapiens 34-38 16972258-0 2006 N-(4-hydroxyphenyl)retinamide induces apoptosis in human retinal pigment epithelial cells: retinoic acid receptors regulate apoptosis, reactive oxygen species generation, and the expression of heme oxygenase-1 and Gadd153. Fenretinide 0-29 heme oxygenase 1 Homo sapiens 193-209 17020887-5 2006 These effects were blocked by the HO-1 inhibitors Zn- and Sn-protoporphyrin IX. Tin complex of Protoporphyrin-IX 58-78 heme oxygenase 1 Homo sapiens 34-38 16972258-7 2006 In addition, the increase in the expression of heme oxygenase-1 (HO-1), a stress response protein, and the growth arrest and DNA damage-inducible transcription factor 153 (Gadd153) in response to the ROS generation were also blocked by these receptor antagonists. Reactive Oxygen Species 200-203 heme oxygenase 1 Homo sapiens 47-63 17020887-7 2006 While increased HO-1 activity prevented NO-induced fluctuations in transition metal homeostasis, addition of an iron chelator decreased NO toxicity only slightly. Metals 78-83 heme oxygenase 1 Homo sapiens 16-20 17020887-8 2006 Our results indicate that lipid peroxidation is a significant cause of NO-induced necrosis in human lung epithelial cells, and that the increased NO survival of L1 cells is due at least in part to decreased lipid peroxidation mediated by HO-1-generated biliverdin or bilirubin. Biliverdine 253-263 heme oxygenase 1 Homo sapiens 238-242 17020887-8 2006 Our results indicate that lipid peroxidation is a significant cause of NO-induced necrosis in human lung epithelial cells, and that the increased NO survival of L1 cells is due at least in part to decreased lipid peroxidation mediated by HO-1-generated biliverdin or bilirubin. Bilirubin 267-276 heme oxygenase 1 Homo sapiens 238-242 17027882-6 2006 According to our data, sodium nitroprusside (SNP) increased HO-1 expression in a concentration dependent manner. Nitroprusside 23-43 heme oxygenase 1 Homo sapiens 60-64 17199800-0 2006 Carnitine"S protective effect on oxidative stress is mediated by heme oxygenase-1. Carnitine 0-9 heme oxygenase 1 Homo sapiens 65-81 17138185-0 2006 Heme oxygenase-1 mediates cytoprotection against nitric oxide-induced cytotoxicity via the cGMP pathway in human pulp cells. Nitric Oxide 49-61 heme oxygenase 1 Homo sapiens 0-16 17138185-0 2006 Heme oxygenase-1 mediates cytoprotection against nitric oxide-induced cytotoxicity via the cGMP pathway in human pulp cells. Cyclic GMP 91-95 heme oxygenase 1 Homo sapiens 0-16 17138185-1 2006 OBJECTIVE: This study examined the effects of exogenous nitric oxide (NO) on human pulp cells and the involvement of cyclic 3",5"-monophosphate (cGMP) in pulpal protection induced by heme oxygenase-1 (HO-1) against NO-induced cytotoxicity. cyclic 3",5"-monophosphate 117-143 heme oxygenase 1 Homo sapiens 183-199 17138185-1 2006 OBJECTIVE: This study examined the effects of exogenous nitric oxide (NO) on human pulp cells and the involvement of cyclic 3",5"-monophosphate (cGMP) in pulpal protection induced by heme oxygenase-1 (HO-1) against NO-induced cytotoxicity. cyclic 3",5"-monophosphate 117-143 heme oxygenase 1 Homo sapiens 201-205 17138185-1 2006 OBJECTIVE: This study examined the effects of exogenous nitric oxide (NO) on human pulp cells and the involvement of cyclic 3",5"-monophosphate (cGMP) in pulpal protection induced by heme oxygenase-1 (HO-1) against NO-induced cytotoxicity. Cyclic GMP 145-149 heme oxygenase 1 Homo sapiens 183-199 17138185-2 2006 STUDY DESIGN: This study investigated cytotoxicity and HO-1 induction in pulp cells induced by the NO donor S-nitroso-N-acetyl-D,L-penicillamine (SNAP), by using Western blotting and a cell viability assay. snap 108-144 heme oxygenase 1 Homo sapiens 55-59 17138185-4 2006 RESULTS: S-nitroso-N-acetyl-D,L-penicillamine decreased cell viability, but increased HO-1 expression in a concentration- and time-dependent manner in human pulp cells. snap 9-45 heme oxygenase 1 Homo sapiens 86-90 17138185-5 2006 NO-induced cytotoxicity was inhibited in the presence of hemin (inducer of HO-1), whereas it was enhanced in the presence of zinc protoporphyrin IX (ZnPP IX, HO-1 inhibitor); therefore, the NO-induced cytotoxicity was correlated with HO-1 expression. zinc protoporphyrin 125-147 heme oxygenase 1 Homo sapiens 158-162 17138185-5 2006 NO-induced cytotoxicity was inhibited in the presence of hemin (inducer of HO-1), whereas it was enhanced in the presence of zinc protoporphyrin IX (ZnPP IX, HO-1 inhibitor); therefore, the NO-induced cytotoxicity was correlated with HO-1 expression. zinc protoporphyrin 125-147 heme oxygenase 1 Homo sapiens 158-162 17138185-6 2006 Pretreatment with a membrane-permeable cGMP analog, 8-bromo-cGMP, restored cell death and enhanced the HO-1 protein expression induced by SNAP. Cyclic GMP 39-43 heme oxygenase 1 Homo sapiens 103-107 17138185-6 2006 Pretreatment with a membrane-permeable cGMP analog, 8-bromo-cGMP, restored cell death and enhanced the HO-1 protein expression induced by SNAP. 8-bromocyclic GMP 52-64 heme oxygenase 1 Homo sapiens 103-107 17138185-8 2006 CONCLUSION: These findings of a link between HO-1, regulated via the cGMP system and NO-induced cytotoxicity in human pulp cells, suggest a protective role for HO-1 in pulpal inflammation. Cyclic GMP 69-73 heme oxygenase 1 Homo sapiens 45-49 17138185-8 2006 CONCLUSION: These findings of a link between HO-1, regulated via the cGMP system and NO-induced cytotoxicity in human pulp cells, suggest a protective role for HO-1 in pulpal inflammation. Cyclic GMP 69-73 heme oxygenase 1 Homo sapiens 160-164 17027882-7 2006 Preconditioning with low concentration SNP (0.3mM) inhibited subsequent high concentration SNP (1.5mM)-induced apoptosis, and this effect was reversed by the HO-1 inhibitor SnPP. S-Nitroso-N-propionyl-D,L-penicillamine 173-177 heme oxygenase 1 Homo sapiens 158-162 16950787-7 2006 Because we observed notably high levels of phosphorylated protein kinase C alpha and its suppression by EGCG and deferoxamine (an iron chelator), a possible mechanism involving phosphorylated protein kinase C alpha and iron in Nrf2-HO-1 activation was further investigated. Iron 219-223 heme oxygenase 1 Homo sapiens 232-236 16950787-8 2006 Collectively, our findings suggest that Nrf2-mediated HO-1 overexpression confers resistance to apoptosis induction by EGCG; therefore, its inactivation may be a target for overcoming the resistance to chemoprevention and chemotherapy. epigallocatechin gallate 119-123 heme oxygenase 1 Homo sapiens 54-58 17049479-6 2006 Oxidised LDL treatment also increased the expression of the antioxidant stress protein heme oxygenase-1 in macrophages; however, this increase was markedly attenuated by ascorbate pretreatment. Ascorbic Acid 170-179 heme oxygenase 1 Homo sapiens 87-103 16950787-0 2006 Constitutive overexpression of Nrf2-dependent heme oxygenase-1 in A549 cells contributes to resistance to apoptosis induced by epigallocatechin 3-gallate. epigallocatechin gallate 127-153 heme oxygenase 1 Homo sapiens 46-62 16950787-4 2006 Because we found that A549 cells constitutively overexpress HO-1 and its associated transcription factor Nrf2, we tested an hypothesis that EGCG resistance in these cells may be linked with Nrf2-mediated HO-1 overexpression. epigallocatechin gallate 140-144 heme oxygenase 1 Homo sapiens 60-64 16950787-4 2006 Because we found that A549 cells constitutively overexpress HO-1 and its associated transcription factor Nrf2, we tested an hypothesis that EGCG resistance in these cells may be linked with Nrf2-mediated HO-1 overexpression. epigallocatechin gallate 140-144 heme oxygenase 1 Homo sapiens 204-208 16950787-5 2006 HO-1 inhibition with tin-protoporphyrin IX and silencing with RNA interference rendered cells more sensitive to apoptosis induction by EGCG and classical prooxidants. tin protoporphyrin IX 21-42 heme oxygenase 1 Homo sapiens 0-4 16950787-5 2006 HO-1 inhibition with tin-protoporphyrin IX and silencing with RNA interference rendered cells more sensitive to apoptosis induction by EGCG and classical prooxidants. epigallocatechin gallate 135-139 heme oxygenase 1 Homo sapiens 0-4 16950787-6 2006 Interestingly, EGCG at high concentration (>200 microm) induced apoptosis by suppressing expression of HO-1 protein and mRNA, and this effect correlated with a decrease in both Nrf2-ARE binding and HO-1-ARE-luciferase activity, suggesting Nrf2-driven transcriptional activation of ho-1. epigallocatechin gallate 15-19 heme oxygenase 1 Homo sapiens 106-110 16950787-6 2006 Interestingly, EGCG at high concentration (>200 microm) induced apoptosis by suppressing expression of HO-1 protein and mRNA, and this effect correlated with a decrease in both Nrf2-ARE binding and HO-1-ARE-luciferase activity, suggesting Nrf2-driven transcriptional activation of ho-1. epigallocatechin gallate 15-19 heme oxygenase 1 Homo sapiens 201-205 16950787-6 2006 Interestingly, EGCG at high concentration (>200 microm) induced apoptosis by suppressing expression of HO-1 protein and mRNA, and this effect correlated with a decrease in both Nrf2-ARE binding and HO-1-ARE-luciferase activity, suggesting Nrf2-driven transcriptional activation of ho-1. epigallocatechin gallate 15-19 heme oxygenase 1 Homo sapiens 284-288 16950787-7 2006 Because we observed notably high levels of phosphorylated protein kinase C alpha and its suppression by EGCG and deferoxamine (an iron chelator), a possible mechanism involving phosphorylated protein kinase C alpha and iron in Nrf2-HO-1 activation was further investigated. epigallocatechin gallate 104-108 heme oxygenase 1 Homo sapiens 232-236 16950787-7 2006 Because we observed notably high levels of phosphorylated protein kinase C alpha and its suppression by EGCG and deferoxamine (an iron chelator), a possible mechanism involving phosphorylated protein kinase C alpha and iron in Nrf2-HO-1 activation was further investigated. Deferoxamine 113-125 heme oxygenase 1 Homo sapiens 232-236 16950787-7 2006 Because we observed notably high levels of phosphorylated protein kinase C alpha and its suppression by EGCG and deferoxamine (an iron chelator), a possible mechanism involving phosphorylated protein kinase C alpha and iron in Nrf2-HO-1 activation was further investigated. Iron 130-134 heme oxygenase 1 Homo sapiens 232-236 16982957-3 2006 In human microvascular endothelial cells, the NO donor DETA-NONOate (0.1 to 1 mmol/L) strongly induced expression of heme oxygenase-1 but not Cu/Zn superoxide dismutase. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 55-67 heme oxygenase 1 Homo sapiens 117-133 17140381-0 2006 Changes in expression of genes encoding antioxidant enzymes, heme oxygenase-1, Bcl-2, and Bcl-xl and in level of reactive oxygen species in tumor cells resistant to doxorubicin. Doxorubicin 165-176 heme oxygenase 1 Homo sapiens 61-77 17140381-1 2006 The relationship between expression of genes encoding key antioxidant enzymes, heme oxygenase-1, Bcl-2, and Bcl-xl and change in production of reactive oxygen species (ROS) resulting from development of resistance of cancer cells K562, MCF-7, and SKOV-3 to the prooxidant chemotherapeutic agent doxorubicin (DOX) has been studied. Reactive Oxygen Species 143-166 heme oxygenase 1 Homo sapiens 79-95 17140381-1 2006 The relationship between expression of genes encoding key antioxidant enzymes, heme oxygenase-1, Bcl-2, and Bcl-xl and change in production of reactive oxygen species (ROS) resulting from development of resistance of cancer cells K562, MCF-7, and SKOV-3 to the prooxidant chemotherapeutic agent doxorubicin (DOX) has been studied. Reactive Oxygen Species 168-171 heme oxygenase 1 Homo sapiens 79-95 16999951-6 2006 RESULTS: HO-1 expression and activity (5.3+/-2.1 nmol bilirubin/mg protein/h) were only present in ECs from advanced atherosclerotic lesions. Bilirubin 54-63 heme oxygenase 1 Homo sapiens 9-13 16982957-4 2006 This was associated with a reduction of the superoxide-generating capacity of NADPH oxidase, an effect that depended on de novo gene transcription and heme oxygenase-1 activity. Superoxides 44-54 heme oxygenase 1 Homo sapiens 151-167 16982957-6 2006 DETA-NONOate alone had little effect on TNF-stimulated reactive oxygen species, but it enhanced the TNF response when: (1) heme oxygenase-1 expression was blocked with specific small-interfering RNA; (2) heme oxygenase-1 activity was blocked by zinc-protoporphyrin; or (3) NADPH oxidase activity was blocked by diphenyleneiodonium. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 0-12 heme oxygenase 1 Homo sapiens 123-139 16982957-6 2006 DETA-NONOate alone had little effect on TNF-stimulated reactive oxygen species, but it enhanced the TNF response when: (1) heme oxygenase-1 expression was blocked with specific small-interfering RNA; (2) heme oxygenase-1 activity was blocked by zinc-protoporphyrin; or (3) NADPH oxidase activity was blocked by diphenyleneiodonium. 2,2'-(hydroxynitrosohydrazono)bis-ethanamine 0-12 heme oxygenase 1 Homo sapiens 204-220 16982957-6 2006 DETA-NONOate alone had little effect on TNF-stimulated reactive oxygen species, but it enhanced the TNF response when: (1) heme oxygenase-1 expression was blocked with specific small-interfering RNA; (2) heme oxygenase-1 activity was blocked by zinc-protoporphyrin; or (3) NADPH oxidase activity was blocked by diphenyleneiodonium. zinc protoporphyrin 245-264 heme oxygenase 1 Homo sapiens 204-220 16982957-6 2006 DETA-NONOate alone had little effect on TNF-stimulated reactive oxygen species, but it enhanced the TNF response when: (1) heme oxygenase-1 expression was blocked with specific small-interfering RNA; (2) heme oxygenase-1 activity was blocked by zinc-protoporphyrin; or (3) NADPH oxidase activity was blocked by diphenyleneiodonium. diphenyleneiodonium 311-330 heme oxygenase 1 Homo sapiens 204-220 16982957-7 2006 Moreover, the heme oxygenase-1 end product bilirubin directly inhibited fully functional NADPH oxidase and seemed to interrupt the assembly and activation of the oxidase. Bilirubin 43-52 heme oxygenase 1 Homo sapiens 14-30 16982957-8 2006 In conclusion, NO may modulate superoxide production by NADPH oxidase in human vascular endothelial cells, at least partly by inducing heme oxygenase-1. Superoxides 31-41 heme oxygenase 1 Homo sapiens 135-151 16982957-9 2006 Our results indicate that suppression of NADPH oxidase-dependent reactive oxygen species formation may represent a novel mechanism underlying the cardiovascular protective actions of heme oxygenase-1 and bilirubin. Reactive Oxygen Species 65-88 heme oxygenase 1 Homo sapiens 183-199 17048120-5 2006 Whereas BHA failed to induce HO-1 in wild-type and Nrf2 knock-out MFs, tBHQ strongly induced HO-1 in wild-type, but not in Nrf2 knock-out MFs. 2-tert-butylhydroquinone 71-75 heme oxygenase 1 Homo sapiens 93-97 17099512-9 2006 Inhibition of HO-1 activity by Zn-PP administration abolished the beneficial effect of hemin-pretreatment. zinc protoporphyrin 31-36 heme oxygenase 1 Homo sapiens 14-18 16739127-4 2006 In this study, we investigated the potential roles of extracellular signal-regulated kinase (ERK) and c-jun N-terminal kinase (JNK) in the regulation of butylated hydroxyanisole (BHA)-induced and Nrf2-dependent ARE transcriptional activity and ARE-mediated endogenous heme oxygenase-1 (HO-1) protein expression in HepG2 cells. Butylated Hydroxyanisole 179-182 heme oxygenase 1 Homo sapiens 268-284 16739127-4 2006 In this study, we investigated the potential roles of extracellular signal-regulated kinase (ERK) and c-jun N-terminal kinase (JNK) in the regulation of butylated hydroxyanisole (BHA)-induced and Nrf2-dependent ARE transcriptional activity and ARE-mediated endogenous heme oxygenase-1 (HO-1) protein expression in HepG2 cells. Butylated Hydroxyanisole 179-182 heme oxygenase 1 Homo sapiens 286-290 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Butylated Hydroxyanisole 109-133 heme oxygenase 1 Homo sapiens 13-29 17048120-6 2006 CONCLUSIONS: Our data demonstrate that both BHA and tBHQ are strong chemical inducers of HO-1, NQO1 and Nrf2 proteins in primary-cultured human and rat hepatocytes with the activation of MAPK ERK1/2 and JNK1/2. Butylated Hydroxyanisole 44-47 heme oxygenase 1 Homo sapiens 89-93 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Butylated Hydroxyanisole 109-133 heme oxygenase 1 Homo sapiens 31-35 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Butylated Hydroxyanisole 135-138 heme oxygenase 1 Homo sapiens 13-29 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. Butylated Hydroxyanisole 135-138 heme oxygenase 1 Homo sapiens 31-35 17048120-6 2006 CONCLUSIONS: Our data demonstrate that both BHA and tBHQ are strong chemical inducers of HO-1, NQO1 and Nrf2 proteins in primary-cultured human and rat hepatocytes with the activation of MAPK ERK1/2 and JNK1/2. 2-tert-butylhydroquinone 52-56 heme oxygenase 1 Homo sapiens 89-93 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. 2-tert-butylhydroquinone 160-182 heme oxygenase 1 Homo sapiens 13-29 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. 2-tert-butylhydroquinone 160-182 heme oxygenase 1 Homo sapiens 31-35 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. 2-tert-butylhydroquinone 184-188 heme oxygenase 1 Homo sapiens 13-29 17048120-0 2006 Induction of heme oxygenase-1 (HO-1) and NAD[P]H: quinone oxidoreductase 1 (NQO1) by a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) in primary-cultured human and rat hepatocytes. 2-tert-butylhydroquinone 184-188 heme oxygenase 1 Homo sapiens 31-35 17048120-1 2006 PURPOSE: This study was aimed to investigate the effects of a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) on the induction of HO-1, NQO1 and Nrf2 proteins and their regulatory mechanisms in primary-cultured hepatocytes. Butylated Hydroxyanisole 84-108 heme oxygenase 1 Homo sapiens 185-189 17048120-1 2006 PURPOSE: This study was aimed to investigate the effects of a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) on the induction of HO-1, NQO1 and Nrf2 proteins and their regulatory mechanisms in primary-cultured hepatocytes. Butylated Hydroxyanisole 110-113 heme oxygenase 1 Homo sapiens 185-189 17048120-1 2006 PURPOSE: This study was aimed to investigate the effects of a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) on the induction of HO-1, NQO1 and Nrf2 proteins and their regulatory mechanisms in primary-cultured hepatocytes. 2-tert-butylhydroquinone 135-157 heme oxygenase 1 Homo sapiens 185-189 17048120-1 2006 PURPOSE: This study was aimed to investigate the effects of a phenolic antioxidant, butylated hydroxyanisole (BHA) and its metabolite, tert-butylhydroquinone (tBHQ) on the induction of HO-1, NQO1 and Nrf2 proteins and their regulatory mechanisms in primary-cultured hepatocytes. 2-tert-butylhydroquinone 159-163 heme oxygenase 1 Homo sapiens 185-189 17048120-7 2006 However, in MFs, BHA failed to induce HO-1, whereas tBHQ strongly induced HO-1 in Nrf2 wild-type but not in Nrf2 knock-out, suggesting that Nrf2 is indispensable for tBHQ-induced HO-1 in MF. 2-tert-butylhydroquinone 52-56 heme oxygenase 1 Homo sapiens 74-78 17048120-7 2006 However, in MFs, BHA failed to induce HO-1, whereas tBHQ strongly induced HO-1 in Nrf2 wild-type but not in Nrf2 knock-out, suggesting that Nrf2 is indispensable for tBHQ-induced HO-1 in MF. 2-tert-butylhydroquinone 52-56 heme oxygenase 1 Homo sapiens 74-78 16858012-0 2006 Heme oxygenase-1, a potential biomarker of chronic silicosis, attenuates silica-induced lung injury. Silicon Dioxide 73-79 heme oxygenase 1 Homo sapiens 0-16 16858012-8 2006 Serum HO-1 levels also correlated inversely with serum 8-hydroxydeoxyguanosine levels and positively with vital capacity and forced expiratory volume in one second in patients with silicosis. 8-ohdg 55-78 heme oxygenase 1 Homo sapiens 6-10 16858012-9 2006 HO-1 was present in the lungs of humans and mice with silicosis, especially at sites of silica particle deposition. Silicon Dioxide 88-94 heme oxygenase 1 Homo sapiens 0-4 16858012-11 2006 The inflammation was suppressed by treatment with hemin, an inducer of HO-1, and enhanced by zinc protoporphyrin, an inhibitor of HO-1. Hemin 50-55 heme oxygenase 1 Homo sapiens 71-75 16858012-11 2006 The inflammation was suppressed by treatment with hemin, an inducer of HO-1, and enhanced by zinc protoporphyrin, an inhibitor of HO-1. zinc protoporphyrin 93-112 heme oxygenase 1 Homo sapiens 130-134 16858012-13 2006 HO-1 suppresses reactive oxygen species activity, and subsequent pathologic changes, thereby attenuating disease progression. Reactive Oxygen Species 16-39 heme oxygenase 1 Homo sapiens 0-4 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Heme 73-77 heme oxygenase 1 Homo sapiens 0-16 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Heme 73-77 heme oxygenase 1 Homo sapiens 18-22 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Biliverdine 83-93 heme oxygenase 1 Homo sapiens 0-16 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Biliverdine 83-93 heme oxygenase 1 Homo sapiens 18-22 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Iron 110-114 heme oxygenase 1 Homo sapiens 0-16 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Iron 110-114 heme oxygenase 1 Homo sapiens 18-22 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Carbon Monoxide 119-134 heme oxygenase 1 Homo sapiens 0-16 17042977-2 2006 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in the catabolism of heme into biliverdin, releasing free iron and carbon monoxide. Carbon Monoxide 119-134 heme oxygenase 1 Homo sapiens 18-22 17042977-9 2006 Flow cytometry was used to detect apoptotic cells in the HO-1 group and in the control group after induction by recombinant human tumor necrosis factor-alpha (rTNFalpha) and cycloheximide (CHX) for 48 hours. Cycloheximide 174-187 heme oxygenase 1 Homo sapiens 57-61 16485034-0 2006 Flunarizine induces Nrf2-mediated transcriptional activation of heme oxygenase-1 in protection of auditory cells from cisplatin. Flunarizine 0-11 heme oxygenase 1 Homo sapiens 64-80 16375908-4 2006 We found that, in vascular endothelial cells, simvastatin increased the steady-state levels of heat shock proteins 90 and 70, and heme oxygenase-1 and caused the nuclear translocation of heat shock factor 1. Simvastatin 46-57 heme oxygenase 1 Homo sapiens 130-146 16953189-5 2006 KEY RESULTS: Intraperitoneal challenge with carrageenan enhanced HO-1 protein expression in mesentery and bilirubin concentration in peritoneal exudates. Carrageenan 44-55 heme oxygenase 1 Homo sapiens 65-69 16953189-6 2006 Pretreatment of mice with a non-specific inhibitor of HO (ZnDPBG) or with a HO-1 specific inhibitor (ZnPP IX) enhanced neutrophil migration, rolling and adhesion on endothelium induced by carrageenan. zinc protoporphyrin 101-108 heme oxygenase 1 Homo sapiens 76-80 16953189-6 2006 Pretreatment of mice with a non-specific inhibitor of HO (ZnDPBG) or with a HO-1 specific inhibitor (ZnPP IX) enhanced neutrophil migration, rolling and adhesion on endothelium induced by carrageenan. Carrageenan 188-199 heme oxygenase 1 Homo sapiens 76-80 16485034-5 2006 Furthermore, both pharmacological inhibition and siRNA transfection of HO-1 completely abolished the flunarizine-mediated protection of HEI-OC1 cells and the primary rat (P2) organ of Corti explants from cisplatin. Cisplatin 204-213 heme oxygenase 1 Homo sapiens 71-75 16485034-0 2006 Flunarizine induces Nrf2-mediated transcriptional activation of heme oxygenase-1 in protection of auditory cells from cisplatin. Cisplatin 118-127 heme oxygenase 1 Homo sapiens 64-80 16485034-6 2006 These results suggest that Nrf2-driven transcriptional activation of ARE through PI3K-Akt signaling augments the generation of HO-1, which may be a critically important determinant in cellular response toward cisplatin and the cytoprotective effect of flunarizine against cisplatin. Cisplatin 209-218 heme oxygenase 1 Homo sapiens 127-131 16485034-6 2006 These results suggest that Nrf2-driven transcriptional activation of ARE through PI3K-Akt signaling augments the generation of HO-1, which may be a critically important determinant in cellular response toward cisplatin and the cytoprotective effect of flunarizine against cisplatin. Flunarizine 252-263 heme oxygenase 1 Homo sapiens 127-131 16485034-6 2006 These results suggest that Nrf2-driven transcriptional activation of ARE through PI3K-Akt signaling augments the generation of HO-1, which may be a critically important determinant in cellular response toward cisplatin and the cytoprotective effect of flunarizine against cisplatin. Cisplatin 272-281 heme oxygenase 1 Homo sapiens 127-131 16485034-4 2006 Pretreatment with flunarizine predominantly increased the transcriptional activity of HO-1 among Nrf2-driven transcripts, including HO-1, NQO1, GCLC, GCLM, GST micro-1, and GSTA4. Flunarizine 18-29 heme oxygenase 1 Homo sapiens 86-90 16485034-4 2006 Pretreatment with flunarizine predominantly increased the transcriptional activity of HO-1 among Nrf2-driven transcripts, including HO-1, NQO1, GCLC, GCLM, GST micro-1, and GSTA4. Flunarizine 18-29 heme oxygenase 1 Homo sapiens 132-136 16485034-5 2006 Furthermore, both pharmacological inhibition and siRNA transfection of HO-1 completely abolished the flunarizine-mediated protection of HEI-OC1 cells and the primary rat (P2) organ of Corti explants from cisplatin. Flunarizine 101-112 heme oxygenase 1 Homo sapiens 71-75 16775837-3 2006 To investigate the mechanisms for this inhibition, Cr(VI) effects on basal and arsenic (As(III))-induced HO-1 expression were examined in cultured human bronchial epithelial (BEAS-2B) cells. Chromium 51-53 heme oxygenase 1 Homo sapiens 105-109 16775837-3 2006 To investigate the mechanisms for this inhibition, Cr(VI) effects on basal and arsenic (As(III))-induced HO-1 expression were examined in cultured human bronchial epithelial (BEAS-2B) cells. Arsenic 79-86 heme oxygenase 1 Homo sapiens 105-109 16775837-3 2006 To investigate the mechanisms for this inhibition, Cr(VI) effects on basal and arsenic (As(III))-induced HO-1 expression were examined in cultured human bronchial epithelial (BEAS-2B) cells. as(iii) 88-95 heme oxygenase 1 Homo sapiens 105-109 16775837-4 2006 An effect of Cr(VI) on the low basal HO-1 mRNA and protein levels in BEAS-2B cells was not detectible. chromium hexavalent ion 13-19 heme oxygenase 1 Homo sapiens 37-41 16951197-2 2006 Using human HO-1 promoter reporter plasmids and ChIP assay, we have identified that sulforaphane transcriptionally activated the upstream ARE-rich enhancer region, located at -9.0 kb upstream human HO-1 promoter. sulforaphane 84-96 heme oxygenase 1 Homo sapiens 198-202 16596642-2 2006 Recently, a (GT)n dinucleotide repeat polymorphism in the HO-1 promoter was shown to modulate HO-1 gene expression. (gt)n dinucleotide 12-30 heme oxygenase 1 Homo sapiens 58-62 16596642-2 2006 Recently, a (GT)n dinucleotide repeat polymorphism in the HO-1 promoter was shown to modulate HO-1 gene expression. (gt)n dinucleotide 12-30 heme oxygenase 1 Homo sapiens 94-98 16840713-0 2006 Protection of human vascular smooth muscle cells from H2O2-induced apoptosis through functional codependence between HO-1 and AKT. Hydrogen Peroxide 54-58 heme oxygenase 1 Homo sapiens 117-129 16840713-9 2006 HO-1 reduced H2O2-induced OS and apoptosis. Hydrogen Peroxide 13-17 heme oxygenase 1 Homo sapiens 0-4 16840713-10 2006 Akt knockdown removed the protective effect of HO-1 on delta psi(m) during exposure to H2O2. Hydrogen Peroxide 87-91 heme oxygenase 1 Homo sapiens 47-51 16840713-12 2006 Inhibition of PI3K-Akt reduced induction of HO-1 protein expression by H2O2 and blocked its anti-apoptotic effects. Hydrogen Peroxide 71-75 heme oxygenase 1 Homo sapiens 44-48 16840713-14 2006 CONCLUSIONS: HO-1 and Akt exert codependent cytoprotective effects against OS-induced apoptosis in HASMCs. hasmcs 99-105 heme oxygenase 1 Homo sapiens 13-17 16951197-0 2006 Mechanism of action of sulforaphane: inhibition of p38 mitogen-activated protein kinase isoforms contributing to the induction of antioxidant response element-mediated heme oxygenase-1 in human hepatoma HepG2 cells. sulforaphane 23-35 heme oxygenase 1 Homo sapiens 168-184 16951197-1 2006 Exposure of sulforaphane to HepG2 cells increased heme oxygenase-1 (HO-1) expression by activating antioxidant response element (ARE) through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). sulforaphane 12-24 heme oxygenase 1 Homo sapiens 50-66 16951197-1 2006 Exposure of sulforaphane to HepG2 cells increased heme oxygenase-1 (HO-1) expression by activating antioxidant response element (ARE) through induction of Nrf2 and suppression of Kelch-like ECH-associated protein 1 (Keap1). sulforaphane 12-24 heme oxygenase 1 Homo sapiens 68-72 16959797-0 2006 The antioxidant role of a reagent, 2",7"-dichlorodihydrofluorescin diacetate, detecting reactive-oxygen species and blocking the induction of heme oxygenase-1 and preventing cytotoxicity. 2",7"-dichlorodihydrofluorescin diacetate 35-76 heme oxygenase 1 Homo sapiens 142-158 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Heme 33-37 heme oxygenase 1 Homo sapiens 0-16 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Biliverdine 43-53 heme oxygenase 1 Homo sapiens 0-16 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Biliverdine 43-53 heme oxygenase 1 Homo sapiens 18-22 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Iron 55-59 heme oxygenase 1 Homo sapiens 0-16 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Iron 55-59 heme oxygenase 1 Homo sapiens 18-22 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Carbon Monoxide 64-66 heme oxygenase 1 Homo sapiens 0-16 16959797-1 2006 Heme oxygenase-1 (HO-1) degrades heme into biliverdin, iron and CO. Carbon Monoxide 64-66 heme oxygenase 1 Homo sapiens 18-22 16959797-6 2006 511, 21-27], and then examined the effect of DCFH-DA on the induction of HO-1 expression by arsenite, cadmium and hemin, which induce oxidative stress and cytotoxicity. diacetyldichlorofluorescein 45-52 heme oxygenase 1 Homo sapiens 73-77 16959797-6 2006 511, 21-27], and then examined the effect of DCFH-DA on the induction of HO-1 expression by arsenite, cadmium and hemin, which induce oxidative stress and cytotoxicity. arsenite 92-100 heme oxygenase 1 Homo sapiens 73-77 16959797-6 2006 511, 21-27], and then examined the effect of DCFH-DA on the induction of HO-1 expression by arsenite, cadmium and hemin, which induce oxidative stress and cytotoxicity. Cadmium 102-109 heme oxygenase 1 Homo sapiens 73-77 16959797-6 2006 511, 21-27], and then examined the effect of DCFH-DA on the induction of HO-1 expression by arsenite, cadmium and hemin, which induce oxidative stress and cytotoxicity. Hemin 114-119 heme oxygenase 1 Homo sapiens 73-77 16959797-7 2006 We found suppression of the arsenite-, cadmium- and hemin-dependent induction of HO-1 with DCFH-DA. arsenite 28-36 heme oxygenase 1 Homo sapiens 81-85 16959797-7 2006 We found suppression of the arsenite-, cadmium- and hemin-dependent induction of HO-1 with DCFH-DA. Cadmium 39-46 heme oxygenase 1 Homo sapiens 81-85 16959797-7 2006 We found suppression of the arsenite-, cadmium- and hemin-dependent induction of HO-1 with DCFH-DA. diacetyldichlorofluorescein 91-98 heme oxygenase 1 Homo sapiens 81-85 16959797-8 2006 The suppression occurred at the transcriptional level since the promoter activity of the Maf-recognition site of the HO-1 gene decreased with the DCFH-DA treatment. diacetyldichlorofluorescein 146-153 heme oxygenase 1 Homo sapiens 117-121 16951197-2 2006 Using human HO-1 promoter reporter plasmids and ChIP assay, we have identified that sulforaphane transcriptionally activated the upstream ARE-rich enhancer region, located at -9.0 kb upstream human HO-1 promoter. sulforaphane 84-96 heme oxygenase 1 Homo sapiens 12-16 16951197-3 2006 Induction of HO-1 by sulforaphane was attenuated by overexpression of mutant Nrf2 plasmid in HepG2 cells and totally abolished in Nrf2 knockout mouse embryonic keratinocytes and fibroblasts. sulforaphane 21-33 heme oxygenase 1 Homo sapiens 13-17 16951197-8 2006 Collectively, our results indicate that transcriptional activation of Nrf2/ARE is critical in sulforaphane-mediated induction of HO-1, which can be modulated in part by the blockade of p38 MAPK signaling pathway. sulforaphane 94-106 heme oxygenase 1 Homo sapiens 129-133 16927154-0 2006 Ketamine-induced gastroprotection during endotoxemia: role of heme-oxygenase-1. Ketamine 0-8 heme oxygenase 1 Homo sapiens 62-78 16927154-4 2006 We hypothesized that ketamine would diminish gastric injury from lipopolysacharide via down-regulation of nuclear factor-kappass, activator protein-1, and inducible nitric oxide synthase, as well as up-regulation of heme-oxygenase-1. Ketamine 21-29 heme oxygenase 1 Homo sapiens 216-232 16927154-5 2006 Ketamine up-regulated heme-oxygenase-1 and attenuated lipopolysacharide-induced changes in gastric nuclear factor-kappass, activator protein-1, and inducible nitric oxide synthase. Ketamine 0-8 heme oxygenase 1 Homo sapiens 22-38 16816389-0 2006 Pescadillo interacts with the cadmium response element of the human heme oxygenase-1 promoter in renal epithelial cells. pescadillo 0-10 heme oxygenase 1 Homo sapiens 68-84 16953118-1 2006 Recently, it has been reported that curcumin, which is known as a potent antioxidant, acts as a non- stressful and non-cytotoxic inducer of the cytoprotective heme oxygenase (HO)-1. Curcumin 36-44 heme oxygenase 1 Homo sapiens 159-180 16802348-10 2006 Moreover, we found that the activation of the ARE and the induction of HO-1 mRNA caused by Apo were suppressed in the presence of the antioxidant N-acetylcysteine and also that Apo produced intracellular reactive oxygen species (ROS), indicating that the low level of ROS produced by Apo may play a critical role in this phenomenon. Acetylcysteine 146-162 heme oxygenase 1 Homo sapiens 71-75 16802348-10 2006 Moreover, we found that the activation of the ARE and the induction of HO-1 mRNA caused by Apo were suppressed in the presence of the antioxidant N-acetylcysteine and also that Apo produced intracellular reactive oxygen species (ROS), indicating that the low level of ROS produced by Apo may play a critical role in this phenomenon. Reactive Oxygen Species 204-227 heme oxygenase 1 Homo sapiens 71-75 16802348-10 2006 Moreover, we found that the activation of the ARE and the induction of HO-1 mRNA caused by Apo were suppressed in the presence of the antioxidant N-acetylcysteine and also that Apo produced intracellular reactive oxygen species (ROS), indicating that the low level of ROS produced by Apo may play a critical role in this phenomenon. Reactive Oxygen Species 229-232 heme oxygenase 1 Homo sapiens 71-75 16802348-10 2006 Moreover, we found that the activation of the ARE and the induction of HO-1 mRNA caused by Apo were suppressed in the presence of the antioxidant N-acetylcysteine and also that Apo produced intracellular reactive oxygen species (ROS), indicating that the low level of ROS produced by Apo may play a critical role in this phenomenon. Reactive Oxygen Species 268-271 heme oxygenase 1 Homo sapiens 71-75 16816389-0 2006 Pescadillo interacts with the cadmium response element of the human heme oxygenase-1 promoter in renal epithelial cells. Cadmium 30-37 heme oxygenase 1 Homo sapiens 68-84 16816389-3 2006 Exposure of renal proximal tubular epithelial cells to 10 mum cadmium demonstrated induction ( approximately 20-fold) of heme oxygenase-1 mRNA and protein. Cadmium 62-69 heme oxygenase 1 Homo sapiens 121-137 16816389-4 2006 Using a 4.5-kb human heme oxygenase-1 promoter construct, the importance of a previously identified cadmium response element (TGCTAGAT) in HeLa cells was verified in renal epithelial cells. Cadmium 100-107 heme oxygenase 1 Homo sapiens 21-37 16816389-10 2006 The findings demonstrate the important and previously unrecognized role of pescadillo as a DNA-binding protein interacting specifically with the cadmium response element of the human heme oxygenase-1 gene. Cadmium 145-152 heme oxygenase 1 Homo sapiens 183-199 16857833-0 2006 The ACE inhibitory dipeptide Met-Tyr diminishes free radical formation in human endothelial cells via induction of heme oxygenase-1 and ferritin. Dipeptides 19-28 heme oxygenase 1 Homo sapiens 115-131 16867247-9 2006 After the treatment with NaHS for 11 weeks, H(2)S donor ameliorated PVSR and downregulated PCNA expression and ERK activation with an increase in lung tissue CO production and HO-1 protein expression but a decrease in NO production, NOS activity and eNOS protein expression in shunted rats. sodium bisulfide 25-29 heme oxygenase 1 Homo sapiens 176-180 16867247-9 2006 After the treatment with NaHS for 11 weeks, H(2)S donor ameliorated PVSR and downregulated PCNA expression and ERK activation with an increase in lung tissue CO production and HO-1 protein expression but a decrease in NO production, NOS activity and eNOS protein expression in shunted rats. Hydrogen Sulfide 44-49 heme oxygenase 1 Homo sapiens 176-180 16780547-4 2006 Under this scheme, the viability of PEGylated islets was improved; that is, PEG molecules could block cellular immunity and HO-1 could exert its cytoprotective property against inflammation. Polyethylene Glycols 36-39 heme oxygenase 1 Homo sapiens 124-128 16780547-5 2006 Interestingly, when employed with a low dose of CsA (1 mg/kg/day), a cooperative action of PEG molecules and HO-1 in immune reactions could result in the complete survival of transplanted islets for 100 days without islet function impairment. Cyclosporine 48-51 heme oxygenase 1 Homo sapiens 109-113 16246346-0 2006 3-Hydroxyanthranilic acid, one of L-tryptophan metabolites, inhibits monocyte chemoattractant protein-1 secretion and vascular cell adhesion molecule-1 expression via heme oxygenase-1 induction in human umbilical vein endothelial cells. 3-Hydroxyanthranilic Acid 0-25 heme oxygenase 1 Homo sapiens 167-183 16849502-2 2006 This effect is mimicked by CO, generated via the catabolism of heme by HO-1. Carbon Monoxide 27-29 heme oxygenase 1 Homo sapiens 71-75 16849502-2 2006 This effect is mimicked by CO, generated via the catabolism of heme by HO-1. Heme 63-67 heme oxygenase 1 Homo sapiens 71-75 16849502-5 2006 When overexpressed in EC, HO-1 targeted specifically the p38alpha but not the p38beta MAPK isoform for degradation by the 26S proteasome, an effect reversed by the 26S proteasome inhibitors MG-132 or lactacystin. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 190-196 heme oxygenase 1 Homo sapiens 26-30 16849502-5 2006 When overexpressed in EC, HO-1 targeted specifically the p38alpha but not the p38beta MAPK isoform for degradation by the 26S proteasome, an effect reversed by the 26S proteasome inhibitors MG-132 or lactacystin. lactacystin 200-211 heme oxygenase 1 Homo sapiens 26-30 16849502-6 2006 Inhibition of p38alpha expression was also observed when HO-1 was induced physiologically by iron protoporphyrin IX (hemin). iron protoporphyrin IX 93-115 heme oxygenase 1 Homo sapiens 57-61 16849502-9 2006 The antiapoptotic effect of HO-1 was impaired when p38alpha expression was restored ectopically or when its degradation by the 26S proteasome was inhibited by MG-132. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 159-165 heme oxygenase 1 Homo sapiens 28-32 16857833-0 2006 The ACE inhibitory dipeptide Met-Tyr diminishes free radical formation in human endothelial cells via induction of heme oxygenase-1 and ferritin. H-MET-TYR-OH 29-36 heme oxygenase 1 Homo sapiens 115-131 16857833-3 2006 The present study investigates the effect of Met-Tyr on the expression of the antioxidant stress proteins, heme oxygenase-1 (HO-1) and ferritin, in endothelial cells derived from the human umbilical vein and their contribution to the decrease in radical formation that occurs under the influence of this dipeptide. H-MET-TYR-OH 45-52 heme oxygenase 1 Homo sapiens 107-123 16857833-3 2006 The present study investigates the effect of Met-Tyr on the expression of the antioxidant stress proteins, heme oxygenase-1 (HO-1) and ferritin, in endothelial cells derived from the human umbilical vein and their contribution to the decrease in radical formation that occurs under the influence of this dipeptide. H-MET-TYR-OH 45-52 heme oxygenase 1 Homo sapiens 125-129 16857833-3 2006 The present study investigates the effect of Met-Tyr on the expression of the antioxidant stress proteins, heme oxygenase-1 (HO-1) and ferritin, in endothelial cells derived from the human umbilical vein and their contribution to the decrease in radical formation that occurs under the influence of this dipeptide. Dipeptides 304-313 heme oxygenase 1 Homo sapiens 107-123 16857833-5 2006 This indirect protection was associated with a significant increase in protein expression of HO-1 and ferritin and abolished by the HO inhibitor zinc deuteroporphyrin IX 2,4-bis-ethylene glycol (ZnBG). znbg 195-199 heme oxygenase 1 Homo sapiens 93-97 16857833-7 2006 Met-Tyr raised HO-1 mRNA levels by enhancing mRNA stability. H-MET-TYR-OH 0-7 heme oxygenase 1 Homo sapiens 15-19 16857833-9 2006 Our results demonstrate that Met-Tyr protects endothelial cells from oxidative stress via induction of HO-1 and ferritin but independently of its ACE inhibitory properties. H-MET-TYR-OH 29-36 heme oxygenase 1 Homo sapiens 103-107 16480751-0 2006 Upregulation of endothelial heme oxygenase-1 expression through the activation of the JNK pathway by sublethal concentrations of acrolein. Acrolein 129-137 heme oxygenase 1 Homo sapiens 28-44 16866988-9 2006 In addition, FeSO(4) reversed the protective effect of DFO on DA-induced cytotoxicity, HO-1 expression and caspase-3 activation. ferrous sulfate 13-20 heme oxygenase 1 Homo sapiens 87-91 16866988-9 2006 In addition, FeSO(4) reversed the protective effect of DFO on DA-induced cytotoxicity, HO-1 expression and caspase-3 activation. Deferoxamine 55-58 heme oxygenase 1 Homo sapiens 87-91 16928811-3 2006 In the present study, we investigated the roles of extracellular signal-regulated kinase (ERK) and c-Jun-NH(2)-kinase (JNK) in the regulation of phenethyl isothiocyanate (PEITC)-induced and Nrf2-dependent ARE activity and ARE-driven heme oxygenase-1 (HO-1) gene expression in PC-3 cells. phenethyl isothiocyanate 145-169 heme oxygenase 1 Homo sapiens 233-249 16928811-3 2006 In the present study, we investigated the roles of extracellular signal-regulated kinase (ERK) and c-Jun-NH(2)-kinase (JNK) in the regulation of phenethyl isothiocyanate (PEITC)-induced and Nrf2-dependent ARE activity and ARE-driven heme oxygenase-1 (HO-1) gene expression in PC-3 cells. phenethyl isothiocyanate 145-169 heme oxygenase 1 Homo sapiens 251-255 16928811-4 2006 ARE activity and HO-1 expression were strongly increased after treatment with PEITC. phenethyl isothiocyanate 78-83 heme oxygenase 1 Homo sapiens 17-21 16480751-3 2006 In this study, the regulatory effects of acrolein upon the expression of HO-1 were investigated in endothelial cells (ECs). Acrolein 41-49 heme oxygenase 1 Homo sapiens 73-77 16480751-4 2006 We demonstrate that acrolein induces the elevation of HO-1 protein levels, and subsequent enzyme activity, at non-cytotoxic concentrations. Acrolein 20-28 heme oxygenase 1 Homo sapiens 54-58 16480751-5 2006 An additional alpha,beta-unsaturated aldehyde, cinnamaldehyde, was also found to increase HO-1 expression and have less cytotoxicity than acrolein. alpha,beta-unsaturated aldehyde 14-45 heme oxygenase 1 Homo sapiens 90-94 16480751-5 2006 An additional alpha,beta-unsaturated aldehyde, cinnamaldehyde, was also found to increase HO-1 expression and have less cytotoxicity than acrolein. cinnamaldehyde 47-61 heme oxygenase 1 Homo sapiens 90-94 16829200-2 2006 Carbon monoxide is one of the products of the degradation of heme by heme oxygenase 1. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 69-85 16480751-6 2006 Moreover, acrolein-mediated HO-1 induction is abrogated in the presence of actinomycin D and cycloheximide. Dactinomycin 75-88 heme oxygenase 1 Homo sapiens 28-32 16480751-6 2006 Moreover, acrolein-mediated HO-1 induction is abrogated in the presence of actinomycin D and cycloheximide. Cycloheximide 93-106 heme oxygenase 1 Homo sapiens 28-32 16480751-15 2006 In addition, only the JNK inhibitor SP600125 and tyrosine kinase inhibitor genistein had any significant inhibitory impact upon the upregulation of HO-1 by acrolein. pyrazolanthrone 36-44 heme oxygenase 1 Homo sapiens 148-152 16480751-15 2006 In addition, only the JNK inhibitor SP600125 and tyrosine kinase inhibitor genistein had any significant inhibitory impact upon the upregulation of HO-1 by acrolein. Genistein 75-84 heme oxygenase 1 Homo sapiens 148-152 16480751-15 2006 In addition, only the JNK inhibitor SP600125 and tyrosine kinase inhibitor genistein had any significant inhibitory impact upon the upregulation of HO-1 by acrolein. Acrolein 156-164 heme oxygenase 1 Homo sapiens 148-152 16480751-17 2006 Hence, we show in our current experiments that a sublethal concentration of acrolein is in fact a novel HO-1 inducer, and we further identify the principal underlying mechanisms involved in this process. Acrolein 76-84 heme oxygenase 1 Homo sapiens 104-108 16814105-8 2006 Inhibition of HO-1 with Sn protoporphyrin IX and blockage of Sp-1-mediatied upregulation of HO-1 with mithramycin attenuated antioxidant and cytoprotective functions of NGF against hydrogen peroxide. sn protoporphyrin ix 24-44 heme oxygenase 1 Homo sapiens 14-18 16627864-0 2006 Dobutamine improves liver function after hemorrhagic shock through induction of heme oxygenase-1. Dobutamine 0-10 heme oxygenase 1 Homo sapiens 80-96 16814105-8 2006 Inhibition of HO-1 with Sn protoporphyrin IX and blockage of Sp-1-mediatied upregulation of HO-1 with mithramycin attenuated antioxidant and cytoprotective functions of NGF against hydrogen peroxide. Plicamycin 102-113 heme oxygenase 1 Homo sapiens 92-96 16814105-8 2006 Inhibition of HO-1 with Sn protoporphyrin IX and blockage of Sp-1-mediatied upregulation of HO-1 with mithramycin attenuated antioxidant and cytoprotective functions of NGF against hydrogen peroxide. Hydrogen Peroxide 181-198 heme oxygenase 1 Homo sapiens 92-96 16804400-8 2006 Interestingly, gliotoxin induced HO-1 in HT-29 cells and, in turn, inhibition of HO-1 activity by a zinc protoporphyrin IX reversed the effects of gliotoxin in terms of I-kappaB degradation, intercellular adhesion molecule-1 expression, and IL-8 production. Gliotoxin 15-24 heme oxygenase 1 Homo sapiens 33-37 16712795-3 2006 This effect was abrogated in the presence of the heme oxygenase-1 (HO-1) inhibitor ZnBG. zinc deuteroporphyrin IX 2,4-bis(glycol) 83-87 heme oxygenase 1 Homo sapiens 49-65 16712795-3 2006 This effect was abrogated in the presence of the heme oxygenase-1 (HO-1) inhibitor ZnBG. zinc deuteroporphyrin IX 2,4-bis(glycol) 83-87 heme oxygenase 1 Homo sapiens 67-71 16713997-7 2006 Furthermore, l-arginine-induced NO formation was accompanied by a reduction in oxidative stress and an increase in protein expression and enzyme activity of heme oxygenase (HO)-1. Arginine 13-23 heme oxygenase 1 Homo sapiens 157-178 16787441-2 2006 Heme oxygenase consists of two structurally related isozymes, heme oxygenase-1 and and heme oxygenase-2, each of which cleaves heme to form biliverdin, iron and carbon monoxide. Carbon Monoxide 161-176 heme oxygenase 1 Homo sapiens 62-78 16787441-2 2006 Heme oxygenase consists of two structurally related isozymes, heme oxygenase-1 and and heme oxygenase-2, each of which cleaves heme to form biliverdin, iron and carbon monoxide. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 62-78 16787441-2 2006 Heme oxygenase consists of two structurally related isozymes, heme oxygenase-1 and and heme oxygenase-2, each of which cleaves heme to form biliverdin, iron and carbon monoxide. Iron 152-156 heme oxygenase 1 Homo sapiens 62-78 16804400-8 2006 Interestingly, gliotoxin induced HO-1 in HT-29 cells and, in turn, inhibition of HO-1 activity by a zinc protoporphyrin IX reversed the effects of gliotoxin in terms of I-kappaB degradation, intercellular adhesion molecule-1 expression, and IL-8 production. Gliotoxin 15-24 heme oxygenase 1 Homo sapiens 81-85 16804400-8 2006 Interestingly, gliotoxin induced HO-1 in HT-29 cells and, in turn, inhibition of HO-1 activity by a zinc protoporphyrin IX reversed the effects of gliotoxin in terms of I-kappaB degradation, intercellular adhesion molecule-1 expression, and IL-8 production. zinc protoporphyrin 100-122 heme oxygenase 1 Homo sapiens 81-85 16804400-8 2006 Interestingly, gliotoxin induced HO-1 in HT-29 cells and, in turn, inhibition of HO-1 activity by a zinc protoporphyrin IX reversed the effects of gliotoxin in terms of I-kappaB degradation, intercellular adhesion molecule-1 expression, and IL-8 production. Gliotoxin 147-156 heme oxygenase 1 Homo sapiens 33-37 16799064-7 2006 Finally, the ability of flavonoids to induce phase-2 detoxifying enzymes was tested by immunoblot analysis for the transcription factor Nrf2 and the phase-2 gene product heme-oxygenase 1. Flavonoids 24-34 heme oxygenase 1 Homo sapiens 170-186 16799064-13 2006 Many of these flavonoids induced the expression of Nrf2 and the phase-2 gene product heme-oxygenase 1 in human RPE cells. Flavonoids 14-24 heme oxygenase 1 Homo sapiens 85-101 16804400-8 2006 Interestingly, gliotoxin induced HO-1 in HT-29 cells and, in turn, inhibition of HO-1 activity by a zinc protoporphyrin IX reversed the effects of gliotoxin in terms of I-kappaB degradation, intercellular adhesion molecule-1 expression, and IL-8 production. Gliotoxin 147-156 heme oxygenase 1 Homo sapiens 81-85 16624973-0 2006 Nuclear factor-kappa B-independent anti-inflammatory action of salicylate in human endothelial cells: induction of heme oxygenase-1 by the c-jun N-terminal kinase/activator protein-1 pathway. Salicylates 63-73 heme oxygenase 1 Homo sapiens 115-131 16530877-6 2006 Addition of increasing concentrations of N-acetylcysteine (NAC) led to down-regulation of HO-1 in cells expressing HCV proteins. Acetylcysteine 41-57 heme oxygenase 1 Homo sapiens 90-94 16530877-6 2006 Addition of increasing concentrations of N-acetylcysteine (NAC) led to down-regulation of HO-1 in cells expressing HCV proteins. Acetylcysteine 59-62 heme oxygenase 1 Homo sapiens 90-94 16530877-8 2006 Sodium arsenite, a strong inducer of oxidative stress and HO-1, reduced Bach1 expression in wild type Huh-7 cells, and NAC partially abrogated this decrease. sodium arsenite 0-15 heme oxygenase 1 Homo sapiens 58-62 16624973-11 2006 In summary, our study introduces HO-1 as novel NF-kappaB-independent anti-inflammatory target of salicylate in human endothelial cells. Salicylates 97-107 heme oxygenase 1 Homo sapiens 33-37 16624973-6 2006 As judged by Western blot analysis, salicylate increased endothelial heme oxygenase-1 (HO-1) protein levels. Salicylates 36-46 heme oxygenase 1 Homo sapiens 69-85 16624973-12 2006 Moreover, we elucidated the JNK/AP-1 pathway as crucial for the induction of HO-1 by salicylate. Salicylates 85-95 heme oxygenase 1 Homo sapiens 77-81 16624973-6 2006 As judged by Western blot analysis, salicylate increased endothelial heme oxygenase-1 (HO-1) protein levels. Salicylates 36-46 heme oxygenase 1 Homo sapiens 87-91 16624973-7 2006 Using both the HO-1 inhibitor tin(II) protoporphyrin IX and HO-1 antisense oligonucleotides, we causally linked the induction of HO-1 to the decrease of P-selectin. Oligonucleotides 75-91 heme oxygenase 1 Homo sapiens 60-64 16624973-7 2006 Using both the HO-1 inhibitor tin(II) protoporphyrin IX and HO-1 antisense oligonucleotides, we causally linked the induction of HO-1 to the decrease of P-selectin. Oligonucleotides 75-91 heme oxygenase 1 Homo sapiens 60-64 16624973-8 2006 Moreover, we were interested in the signaling mechanisms leading to the up-regulation of HO-1 by salicylate. Salicylates 97-107 heme oxygenase 1 Homo sapiens 89-93 16624973-9 2006 c-Jun NH2-terminal kinase (JNK) was found to be activated by salicylate, and we could causally link this activation to the induction of HO-1 by using the JNK inhibitor 1,9-pyrazoloanthrone. Salicylates 61-71 heme oxygenase 1 Homo sapiens 136-140 16624973-9 2006 c-Jun NH2-terminal kinase (JNK) was found to be activated by salicylate, and we could causally link this activation to the induction of HO-1 by using the JNK inhibitor 1,9-pyrazoloanthrone. pyrazolanthrone 168-188 heme oxygenase 1 Homo sapiens 136-140 16759310-11 2006 In contrast, HO-1 gene expression was correlated negatively with p22phox, TBARS, PCO, HbA(1c) and diabetes duration. tbars 74-79 heme oxygenase 1 Homo sapiens 13-17 16462769-7 2006 On the other hand, zinc (II) protoporphyrin IX, an HO-1 inhibitor, markedly augmented PDT-mediated cytotoxicity towards C-26 and human ovarian carcinoma MDAH2774 cells. zinc;3-[7,12-bis(ethenyl)-18-(3-hydroperoxyprop-1-en-2-yl)-3,8,13,17-tetramethylporphyrin-21,23-diid-2-yl]propanoic acid 19-46 heme oxygenase 1 Homo sapiens 51-55 16462769-7 2006 On the other hand, zinc (II) protoporphyrin IX, an HO-1 inhibitor, markedly augmented PDT-mediated cytotoxicity towards C-26 and human ovarian carcinoma MDAH2774 cells. C-26 120-124 heme oxygenase 1 Homo sapiens 51-55 16462769-8 2006 Neither bilirubin, biliverdin nor carbon monoxide, direct products of HO-1 catalysed heme degradation, was responsible for cytoprotection. Heme 85-89 heme oxygenase 1 Homo sapiens 70-74 16271489-0 2006 Expression of heme oxygenase-1 due to intracellular reactive oxygen species induced by ultrasound. Reactive Oxygen Species 52-75 heme oxygenase 1 Homo sapiens 14-30 16271489-8 2006 These results indicate that superoxide secondarily generated from damaged mitochondria, not hydroxyl radicals generated in medium directly by sonication, give rise to intracellular oxidative stress inducing HO-1 expression. Superoxides 28-38 heme oxygenase 1 Homo sapiens 207-211 16595661-1 2006 Up-regulation of heme oxygenase (HO-1) by either cobalt protoporphyrin (CoPP) or human gene transfer improves vascular and renal function by several mechanisms, including increases in antioxidant levels and decreases in reactive oxygen species (ROS) in vascular and renal tissue. cobaltiprotoporphyrin 49-70 heme oxygenase 1 Homo sapiens 33-37 16595661-1 2006 Up-regulation of heme oxygenase (HO-1) by either cobalt protoporphyrin (CoPP) or human gene transfer improves vascular and renal function by several mechanisms, including increases in antioxidant levels and decreases in reactive oxygen species (ROS) in vascular and renal tissue. cobaltiprotoporphyrin 72-76 heme oxygenase 1 Homo sapiens 33-37 16595661-1 2006 Up-regulation of heme oxygenase (HO-1) by either cobalt protoporphyrin (CoPP) or human gene transfer improves vascular and renal function by several mechanisms, including increases in antioxidant levels and decreases in reactive oxygen species (ROS) in vascular and renal tissue. Reactive Oxygen Species 220-243 heme oxygenase 1 Homo sapiens 33-37 16595661-1 2006 Up-regulation of heme oxygenase (HO-1) by either cobalt protoporphyrin (CoPP) or human gene transfer improves vascular and renal function by several mechanisms, including increases in antioxidant levels and decreases in reactive oxygen species (ROS) in vascular and renal tissue. Reactive Oxygen Species 245-248 heme oxygenase 1 Homo sapiens 33-37 16595661-8 2006 Human HO-1 cDNA transfer into diabetic rats increased both HO-1 protein and activity, and restored mitochondrial ADP/ATP and deoxynucleotide carriers. Adenosine Diphosphate 113-116 heme oxygenase 1 Homo sapiens 6-10 16595661-8 2006 Human HO-1 cDNA transfer into diabetic rats increased both HO-1 protein and activity, and restored mitochondrial ADP/ATP and deoxynucleotide carriers. Adenosine Triphosphate 117-120 heme oxygenase 1 Homo sapiens 6-10 16678019-0 2006 Heme oxygenase-1 attenuates the cisplatin-induced apoptosis of auditory cells via down-regulation of reactive oxygen species generation. Cisplatin 32-41 heme oxygenase 1 Homo sapiens 0-16 16528678-1 2006 Heme oxygenase (HO)-1 is the inducible isoform of the first and rate-controlling enzyme of heme degradation. Heme 91-95 heme oxygenase 1 Homo sapiens 0-21 16728265-2 2006 In honor of my dear friend and colleague, Prof. Dr. Jon van Rood, I summarize here our work in this area making use of heme oxygenase-1 (HO-1) and the products that degradation of heme by HO-1 generates. Heme 119-123 heme oxygenase 1 Homo sapiens 137-141 16728265-2 2006 In honor of my dear friend and colleague, Prof. Dr. Jon van Rood, I summarize here our work in this area making use of heme oxygenase-1 (HO-1) and the products that degradation of heme by HO-1 generates. Heme 119-123 heme oxygenase 1 Homo sapiens 188-192 16378625-0 2006 Upregulation of heme oxygenase-1 by Epigallocatechin-3-gallate via the phosphatidylinositol 3-kinase/Akt and ERK pathways. epigallocatechin gallate 36-62 heme oxygenase 1 Homo sapiens 16-32 16378625-1 2006 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by various phytochemicals and we examined the ability of Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, to upregulate HO-1 expression in endothelial cells (ECs). epigallocatechin gallate 118-144 heme oxygenase 1 Homo sapiens 0-16 16378625-1 2006 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by various phytochemicals and we examined the ability of Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, to upregulate HO-1 expression in endothelial cells (ECs). epigallocatechin gallate 118-144 heme oxygenase 1 Homo sapiens 18-22 16678019-0 2006 Heme oxygenase-1 attenuates the cisplatin-induced apoptosis of auditory cells via down-regulation of reactive oxygen species generation. Reactive Oxygen Species 101-124 heme oxygenase 1 Homo sapiens 0-16 16678019-3 2006 Herein, we demonstrate that pharmacologic induction of HO-1 along with catalytic activation significantly suppressed apoptosis of HEI-OC1 cells induced by cisplatin. Cisplatin 155-164 heme oxygenase 1 Homo sapiens 55-59 16678019-4 2006 Studies of ectopic expression of pcDNA3-HO-1 and siRNA of HO-1 further revealed the protective role of HO-1 against cisplatin in HEI-OC1 cells. Cisplatin 116-125 heme oxygenase 1 Homo sapiens 58-62 16678019-4 2006 Studies of ectopic expression of pcDNA3-HO-1 and siRNA of HO-1 further revealed the protective role of HO-1 against cisplatin in HEI-OC1 cells. Cisplatin 116-125 heme oxygenase 1 Homo sapiens 58-62 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 35-39 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Carbon Monoxide 46-61 heme oxygenase 1 Homo sapiens 130-134 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Carbon Monoxide 63-65 heme oxygenase 1 Homo sapiens 35-39 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Carbon Monoxide 63-65 heme oxygenase 1 Homo sapiens 130-134 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Bilirubin 71-80 heme oxygenase 1 Homo sapiens 35-39 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Bilirubin 71-80 heme oxygenase 1 Homo sapiens 130-134 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Cisplatin 143-152 heme oxygenase 1 Homo sapiens 35-39 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Cisplatin 143-152 heme oxygenase 1 Homo sapiens 130-134 16495208-0 2006 The interaction of nitric oxide with distinct hemoglobins differentially amplifies endothelial heme uptake and heme oxygenase-1 expression. Nitric Oxide 19-31 heme oxygenase 1 Homo sapiens 111-127 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Heme 0-4 heme oxygenase 1 Homo sapiens 61-77 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Heme 0-4 heme oxygenase 1 Homo sapiens 79-83 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Carbon Monoxide 101-116 heme oxygenase 1 Homo sapiens 61-77 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Carbon Monoxide 101-116 heme oxygenase 1 Homo sapiens 79-83 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Iron 118-122 heme oxygenase 1 Homo sapiens 61-77 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Iron 118-122 heme oxygenase 1 Homo sapiens 79-83 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Bilirubin 128-137 heme oxygenase 1 Homo sapiens 61-77 16495208-1 2006 Heme is a strong inducer and substrate of the stress protein heme oxygenase-1 (HO-1), which produces carbon monoxide, iron, and bilirubin. Bilirubin 128-137 heme oxygenase 1 Homo sapiens 79-83 16495208-2 2006 We have reported recently that nitric oxide (NO) augments the incorporation of free hemin in endothelial cells, resulting in amplified HO-1 expression and production of bilirubin. Nitric Oxide 31-43 heme oxygenase 1 Homo sapiens 135-139 16495208-2 2006 We have reported recently that nitric oxide (NO) augments the incorporation of free hemin in endothelial cells, resulting in amplified HO-1 expression and production of bilirubin. Hemin 84-89 heme oxygenase 1 Homo sapiens 135-139 16495208-7 2006 In conclusion, our data indicate a novel role for NO in the modulation of heme transport and HO-1 induction in endothelial cells, which may be relevant for hematological disorders characterized by disruption of the heme-NO equilibrium. Heme 215-219 heme oxygenase 1 Homo sapiens 93-97 16378625-1 2006 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by various phytochemicals and we examined the ability of Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, to upregulate HO-1 expression in endothelial cells (ECs). epigallocatechin gallate 118-144 heme oxygenase 1 Homo sapiens 203-207 16378625-1 2006 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by various phytochemicals and we examined the ability of Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, to upregulate HO-1 expression in endothelial cells (ECs). epigallocatechin gallate 146-150 heme oxygenase 1 Homo sapiens 0-16 16378625-1 2006 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme activated by various phytochemicals and we examined the ability of Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, to upregulate HO-1 expression in endothelial cells (ECs). epigallocatechin gallate 146-150 heme oxygenase 1 Homo sapiens 18-22 16378625-2 2006 We demonstrate that EGCG induces HO-1 expression in a concentration- and time-dependent manner. epigallocatechin gallate 20-24 heme oxygenase 1 Homo sapiens 33-37 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 27-31 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 145-149 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. Dactinomycin 75-88 heme oxygenase 1 Homo sapiens 27-31 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. Dactinomycin 75-88 heme oxygenase 1 Homo sapiens 145-149 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. Cycloheximide 93-106 heme oxygenase 1 Homo sapiens 27-31 16378625-3 2006 Furthermore, EGCG-mediated HO-1 induction was abrogated in the presence of actinomycin D and cycloheximide, indicating that this upregulation of HO-1 occurred at the transcriptional level. Cycloheximide 93-106 heme oxygenase 1 Homo sapiens 145-149 16378625-5 2006 Furthermore, EGCG is the most potent inducer of HO-1 expression of the different green tea constituents that we analyzed, but had no detectable cytotoxic effects over the 25-100 microM dosage range. epigallocatechin gallate 13-17 heme oxygenase 1 Homo sapiens 48-52 16378625-7 2006 In addition, we determined that tyrosine kinase is involved in EGCG induction of HO-1 as this is abrogated by genistein. epigallocatechin gallate 63-67 heme oxygenase 1 Homo sapiens 81-85 16378625-7 2006 In addition, we determined that tyrosine kinase is involved in EGCG induction of HO-1 as this is abrogated by genistein. Genistein 110-119 heme oxygenase 1 Homo sapiens 81-85 16378625-9 2006 In addition, pharmacological inhibitors of phosphatidylinositol 3-kinase and MEK1/2, which are upstream of Akt and ERK1/2, respectively, attenuate EGCG-induced HO-1 expression. epigallocatechin gallate 147-151 heme oxygenase 1 Homo sapiens 160-164 16378625-10 2006 On the other hand, pretreatment of these cells with EGCG exerts significant cytoprotective effects against H2O2, suggesting that the induction of HO-1 is an important component in the protection against oxidative stress. epigallocatechin gallate 52-56 heme oxygenase 1 Homo sapiens 146-150 16378625-10 2006 On the other hand, pretreatment of these cells with EGCG exerts significant cytoprotective effects against H2O2, suggesting that the induction of HO-1 is an important component in the protection against oxidative stress. Hydrogen Peroxide 107-111 heme oxygenase 1 Homo sapiens 146-150 16378625-11 2006 Hence, EGCG is a novel phytochemical inducer of HO-1 expression and we further identify the principal underlying mechanisms involved in this process. epigallocatechin gallate 7-11 heme oxygenase 1 Homo sapiens 48-52 16574070-0 2006 Carbon monoxide mediates heme oxygenase 1 induction via Nrf2 activation in hepatoma cells. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 25-41 16574070-7 2006 Taken together, we suggest that CO induces Nrf2 activation via MAPKs signaling pathways, thereby resulting in HO1 expression in HepG2 cells. Carbon Monoxide 32-34 heme oxygenase 1 Homo sapiens 110-113 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Reactive Oxygen Species 175-198 heme oxygenase 1 Homo sapiens 35-39 16678019-5 2006 Among the catabolic metabolites of HO-1, both carbon monoxide (CO) and bilirubin were directly involved in the protective role of HO-1 against cisplatin through inhibition of reactive oxygen species generation. Reactive Oxygen Species 175-198 heme oxygenase 1 Homo sapiens 130-134 16651638-5 2006 Inhibition of ERK activation with MEK inhibitors (PD98059 or U0126) diminished induction of the antioxidant enzyme heme oxygenase-1. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 50-57 heme oxygenase 1 Homo sapiens 115-131 16651615-7 2006 In steatotic livers, preconditioning and geranylgeranylacetone treatment (which are responsible for HO-1 induction) increased protein kinase C activity. geranylgeranylacetone 41-62 heme oxygenase 1 Homo sapiens 100-104 16651638-5 2006 Inhibition of ERK activation with MEK inhibitors (PD98059 or U0126) diminished induction of the antioxidant enzyme heme oxygenase-1. U 0126 61-66 heme oxygenase 1 Homo sapiens 115-131 16651615-8 2006 HO-1 activators (cobalt(III) protoporphyrin IX) protected both liver types. cobaltiprotoporphyrin 17-46 heme oxygenase 1 Homo sapiens 0-4 16607117-5 2006 RECENT FINDINGS: Recent studies indicate glutamine induces heat shock protein-70, HO-1 (heat shock protein-32), and heat shock protein-27 in models of illness/injury. Glutamine 41-50 heme oxygenase 1 Homo sapiens 82-86 16476737-1 2006 Heme oxygenase-1 (HO-1), the inducible enzyme responsible for the rate-limiting step in the heme catabolism, is expressed in AIDS-Kaposi sarcoma (KS) lesions. Heme 92-96 heme oxygenase 1 Homo sapiens 0-16 16737592-0 2006 [Experimental research on antagonistic effect of heme oxygenase-1 on acute mercury chloride induced renal impairment]. calomel 75-91 heme oxygenase 1 Homo sapiens 49-65 16476785-1 2006 Nitric oxide (NO) and NO donors were among the first reported inducers of the tissue-protective protein heme oxygenase-1 (HO-1) with a potential for eventual use in humans. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 104-120 16476785-1 2006 Nitric oxide (NO) and NO donors were among the first reported inducers of the tissue-protective protein heme oxygenase-1 (HO-1) with a potential for eventual use in humans. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 122-126 16476785-2 2006 Besides other clinically established NO releasing drugs, sodium nitroprusside (SNP) has frequently been employed as an experimental tool to explore effects of NO on HO-1 and other biological targets. Nitroprusside 57-77 heme oxygenase 1 Homo sapiens 165-169 16476785-4 2006 (p. 1633) demonstrate that the effects of SNP on expression of HO-1 are mainly due to free iron released from SNP in aqueous solution, whereas NO plays a negligible role, if any, as the mediator of response to SNP. Iron 91-95 heme oxygenase 1 Homo sapiens 63-67 16476785-5 2006 Downstream effects of iron, after being dissociated from SNP, include increases in intracellular cAMP that are causally linked to subsequent phosphorylation of specific MAPK targets and enhanced HO-1 protein levels. Iron 22-26 heme oxygenase 1 Homo sapiens 195-199 16476785-5 2006 Downstream effects of iron, after being dissociated from SNP, include increases in intracellular cAMP that are causally linked to subsequent phosphorylation of specific MAPK targets and enhanced HO-1 protein levels. Cyclic AMP 97-101 heme oxygenase 1 Homo sapiens 195-199 16476737-1 2006 Heme oxygenase-1 (HO-1), the inducible enzyme responsible for the rate-limiting step in the heme catabolism, is expressed in AIDS-Kaposi sarcoma (KS) lesions. Heme 92-96 heme oxygenase 1 Homo sapiens 18-22 16476737-3 2006 In this study we investigated whether the oncogenic G protein-coupled receptor (KSHV-GPCR or vGPCR), one of the key KSHV genes involved in KS development, activated HO-1 expression. ks 80-82 heme oxygenase 1 Homo sapiens 165-169 16476737-5 2006 Moreover, targeted knock-down gene expression of HO-1 by small hairpin RNA and chemical inhibition of HO-1 enzymatic activity by tin protoporphyrin IX (SnPP), impaired vGPCR-induced survival, proliferation, transformation, and vascular endothelial growth factor (VEGF)-A expression. tin protoporphyrin IX 129-150 heme oxygenase 1 Homo sapiens 102-106 16476737-5 2006 Moreover, targeted knock-down gene expression of HO-1 by small hairpin RNA and chemical inhibition of HO-1 enzymatic activity by tin protoporphyrin IX (SnPP), impaired vGPCR-induced survival, proliferation, transformation, and vascular endothelial growth factor (VEGF)-A expression. S-Nitroso-N-propionyl-D,L-penicillamine 152-156 heme oxygenase 1 Homo sapiens 102-106 16476737-8 2006 On the contrary, administration of the HO-1 inducer cobalt protoporphyrin (CoPP) further enhanced vGPCR-induced tumor growth. cobaltiprotoporphyrin 52-73 heme oxygenase 1 Homo sapiens 39-43 16476737-8 2006 On the contrary, administration of the HO-1 inducer cobalt protoporphyrin (CoPP) further enhanced vGPCR-induced tumor growth. cobaltiprotoporphyrin 75-79 heme oxygenase 1 Homo sapiens 39-43 16631521-0 2006 Heme oxygenase-1 contributes to the cytoprotection of alpha-lipoic acid via activation of p44/42 mitogen-activated protein kinase in vascular smooth muscle cells. Thioctic Acid 54-71 heme oxygenase 1 Homo sapiens 0-16 16636651-7 2006 In addition, expression levels of three other important antioxidant genes, heme oxygenase-1, thioredoxin reductase-1, and manganese superoxide dismutase were monitored by real-time RT-PCR following exposure to TBHP. tert-Butylhydroperoxide 210-214 heme oxygenase 1 Homo sapiens 75-91 16636651-12 2006 In addition, TBHP induced the expression of MTIIa, heme oxygenase-1, thioredoxin reductase-1, and MnSOD in both normal and MTIIa over-expressed cell lines. tert-Butylhydroperoxide 13-17 heme oxygenase 1 Homo sapiens 51-67 16631521-4 2006 ALA significantly induced HO-1 expression accompanied by an increase in HO activity in A10 cells. Thioctic Acid 0-3 heme oxygenase 1 Homo sapiens 26-30 16631525-2 2006 We have earlier shown that a novel antioxidant from the bamboo leaves constituent 3-O-caffeoyl-1-methylquinic acid (MCGA3) induces heme oxygenase-1 (HO-1) and protects endothelial cells from ROS-induced endothelial injury. 3-O-caffeoyl-1-methylquinic acid 82-114 heme oxygenase 1 Homo sapiens 131-147 16631525-2 2006 We have earlier shown that a novel antioxidant from the bamboo leaves constituent 3-O-caffeoyl-1-methylquinic acid (MCGA3) induces heme oxygenase-1 (HO-1) and protects endothelial cells from ROS-induced endothelial injury. 3-O-caffeoyl-1-methylquinic acid 82-114 heme oxygenase 1 Homo sapiens 149-153 16631521-7 2006 ALA significantly increased ROS, and this effect was blocked by N-acetyl-cysteine, which also inhibited ALA-induced activation of p44/42 mitogen-activated protein kinase (MAPK) and AP-1, HO-1 expression, and HO activity. Thioctic Acid 0-3 heme oxygenase 1 Homo sapiens 187-191 16631525-7 2006 By employing N-acetylcysteine and GSH biosynthetic enzyme inhibitors as well as prooxidants, hemin and H(2)O(2), we show that a decreased intracellular GSH/GSSG homeostasis, at least in part, may be involved in the MCGA3-mediated phase II gene induction and Nrf2 translocation, although the attenuation of HO-1 expression with SP 600125 supports a partial involvement of JNK signaling. Glutathione 152-155 heme oxygenase 1 Homo sapiens 306-310 16631521-7 2006 ALA significantly increased ROS, and this effect was blocked by N-acetyl-cysteine, which also inhibited ALA-induced activation of p44/42 mitogen-activated protein kinase (MAPK) and AP-1, HO-1 expression, and HO activity. Acetylcysteine 64-81 heme oxygenase 1 Homo sapiens 187-191 16631525-7 2006 By employing N-acetylcysteine and GSH biosynthetic enzyme inhibitors as well as prooxidants, hemin and H(2)O(2), we show that a decreased intracellular GSH/GSSG homeostasis, at least in part, may be involved in the MCGA3-mediated phase II gene induction and Nrf2 translocation, although the attenuation of HO-1 expression with SP 600125 supports a partial involvement of JNK signaling. Glutathione Disulfide 156-160 heme oxygenase 1 Homo sapiens 306-310 16631521-7 2006 ALA significantly increased ROS, and this effect was blocked by N-acetyl-cysteine, which also inhibited ALA-induced activation of p44/42 mitogen-activated protein kinase (MAPK) and AP-1, HO-1 expression, and HO activity. Thioctic Acid 104-107 heme oxygenase 1 Homo sapiens 187-191 16631521-8 2006 These results suggest that ALA induces HO-1 expression through the production of ROS and subsequent activation of the p44/42 MAPK pathway and AP-1 in vascular smooth muscle cells. Thioctic Acid 27-30 heme oxygenase 1 Homo sapiens 39-43 16631521-8 2006 These results suggest that ALA induces HO-1 expression through the production of ROS and subsequent activation of the p44/42 MAPK pathway and AP-1 in vascular smooth muscle cells. Reactive Oxygen Species 81-84 heme oxygenase 1 Homo sapiens 39-43 16631521-9 2006 This study demonstrated that ALA increases the expression of HO-1, a critical cytoprotective molecule, and identified a novel pleiotropic effect of ALA on cardiovascular protection. Thioctic Acid 29-32 heme oxygenase 1 Homo sapiens 61-65 16631522-7 2006 Intracellular free iron determined with the fluorescent probe calcein rose to approximately 160% of the control level 6 h after SPNO, but declined to approximately 70% after 24 h. Immunoblot analyses revealed a rapid early (approximately 2 h post-NO) increase in heme oxygenase-1 level, followed by a gradual (4-20 h post-NO) increase in ferritin. Iron 19-23 heme oxygenase 1 Homo sapiens 263-279 16759519-9 2006 RESULTS: After stimulation of glucose the insulin concentration in the supernatant of the Ad-HO-1 group was 270 mIU/L +/- 89 mIU/L, significantly higher than those of the Ad-EGFP group (189 mIU/L +/- 88 mIU/L) and control group (182 mIU/L +/- 59 mIU/L, both P < 0.05). Glucose 30-37 heme oxygenase 1 Homo sapiens 93-97 16572448-1 2006 OBJECTIVE: To examine the expression and pathogenetic roles of heme oxygenase 1 (HO-1), an inducible heme-degrading enzyme with antiinflammatory properties, in rheumatoid arthritis (RA). Heme 63-67 heme oxygenase 1 Homo sapiens 81-85 16531144-0 2006 The effect of heme oxygenase-1 induction by glutamine on radiation-induced intestinal damage: the effect of heme oxygenase-1 on radiation enteritis. Glutamine 44-53 heme oxygenase 1 Homo sapiens 14-30 16531144-5 2006 The present study was undertaken to investigate the effect of glutamine administration on heme oxygenase-1 (HO-1) expression of the radiation enteritis model. Glutamine 62-71 heme oxygenase 1 Homo sapiens 90-106 16531144-5 2006 The present study was undertaken to investigate the effect of glutamine administration on heme oxygenase-1 (HO-1) expression of the radiation enteritis model. Glutamine 62-71 heme oxygenase 1 Homo sapiens 108-112 16531144-12 2006 Glutamine treatment was associated with increased HO-1 expression, decreased MPO activity, caspase-3 activity, and MDA levels. Glutamine 0-9 heme oxygenase 1 Homo sapiens 50-54 16531144-13 2006 Inhibition of HO-1 activity by Zn-PP completely eliminated the protective effects of glutamine. zn-pp 31-36 heme oxygenase 1 Homo sapiens 14-18 16531144-13 2006 Inhibition of HO-1 activity by Zn-PP completely eliminated the protective effects of glutamine. Glutamine 85-94 heme oxygenase 1 Homo sapiens 14-18 16531144-18 2006 This protective effect is mediated in part by the induction of HO-1 activity because inhibition of Zn-PP resulted in the complete abolishment of the protective effect of glutamine. zn-pp 99-104 heme oxygenase 1 Homo sapiens 63-67 16531144-18 2006 This protective effect is mediated in part by the induction of HO-1 activity because inhibition of Zn-PP resulted in the complete abolishment of the protective effect of glutamine. Glutamine 170-179 heme oxygenase 1 Homo sapiens 63-67 16572448-6 2006 Hemin, auranofin, and HO-1 expression vector induced HO-1 and reduced expression of tumor necrosis factor alpha (TNFalpha) messenger RNA, lipopolysaccharide (LPS)-induced secretion of interleukin-6 (IL-6) and IL-8, and expression of cyclooxygenase 2 in the synovial cell lines. Auranofin 7-16 heme oxygenase 1 Homo sapiens 53-57 16572448-7 2006 Treatment with HO-1-specific siRNA augmented the synthesis of TNFalpha, IL-6, and IL-8 and canceled the suppressive effects of auranofin on TNFalpha secretion. Auranofin 127-136 heme oxygenase 1 Homo sapiens 15-19 16572448-10 2006 The pharmacologic effects of auranofin depend, in part, on the levels of HO-1, suggesting that HO-1 induction is a novel therapeutic strategy for RA. Auranofin 29-38 heme oxygenase 1 Homo sapiens 73-77 16572448-10 2006 The pharmacologic effects of auranofin depend, in part, on the levels of HO-1, suggesting that HO-1 induction is a novel therapeutic strategy for RA. Auranofin 29-38 heme oxygenase 1 Homo sapiens 95-99 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Carbon Monoxide 271-286 heme oxygenase 1 Homo sapiens 75-79 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Heme 48-52 heme oxygenase 1 Homo sapiens 95-111 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Heme 48-52 heme oxygenase 1 Homo sapiens 113-117 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Carbon Monoxide 122-137 heme oxygenase 1 Homo sapiens 95-111 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Carbon Monoxide 122-137 heme oxygenase 1 Homo sapiens 113-117 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Iron 139-143 heme oxygenase 1 Homo sapiens 95-111 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Iron 139-143 heme oxygenase 1 Homo sapiens 113-117 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Biliverdine 149-159 heme oxygenase 1 Homo sapiens 95-111 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Biliverdine 149-159 heme oxygenase 1 Homo sapiens 113-117 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Bilirubin 185-194 heme oxygenase 1 Homo sapiens 95-111 16545694-1 2006 BACKGROUND: Skin injury leads to the release of heme, a potent prooxidant which is degraded by heme oxygenase-1 (HO-1) to carbon monoxide, iron, and biliverdin, subsequently reduced to bilirubin. Bilirubin 185-194 heme oxygenase 1 Homo sapiens 113-117 16545694-3 2006 RESULTS: Treatment of HaCaT keratinocytes with hemin (heme chloride) induced HO-1 expression and activity. Hemin 47-52 heme oxygenase 1 Homo sapiens 77-81 16545694-3 2006 RESULTS: Treatment of HaCaT keratinocytes with hemin (heme chloride) induced HO-1 expression and activity. heme chloride 54-67 heme oxygenase 1 Homo sapiens 77-81 16545694-5 2006 The involvement of HO-1 in VEGF synthesis was confirmed by inhibition of VEGF expression by SnPPIX, a blocker of HO activity and by attenuation of HO-1 mRNA expression with specific siRNA. tin protoporphyrin IX 92-98 heme oxygenase 1 Homo sapiens 19-23 16545694-6 2006 Importantly, induction of HO-1 by hemin was able to overcome the inhibitory effect of high glucose on VEGF synthesis. Glucose 91-98 heme oxygenase 1 Homo sapiens 26-30 16545694-7 2006 Moreover, HO-1 expression was also induced in keratinocytes cultured in hypoxia, with concomitant augmentation of VEGF production, which was further potentiated by hemin stimulation. Hemin 164-169 heme oxygenase 1 Homo sapiens 10-14 16545694-9 2006 CONCLUSIONS: HO-1 is involved in hemin-induced VEGF expression in HaCaT and may play a role in hypoxic regulation of this protein. Hemin 33-38 heme oxygenase 1 Homo sapiens 13-17 16547262-7 2006 Suppression of HIV replication in hemin-activated cells correlated with the induction of HO-1 and was attenuated by tin protoporphyrin (SnPP) IX, an inhibitor of HO-1 activity, suggesting a pivotal role of this endogenous enzyme in the regulation of HIV infection. tin protoporphyrin IX 116-134 heme oxygenase 1 Homo sapiens 162-166 16547262-7 2006 Suppression of HIV replication in hemin-activated cells correlated with the induction of HO-1 and was attenuated by tin protoporphyrin (SnPP) IX, an inhibitor of HO-1 activity, suggesting a pivotal role of this endogenous enzyme in the regulation of HIV infection. S-Nitroso-N-propionyl-D,L-penicillamine 136-140 heme oxygenase 1 Homo sapiens 162-166 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Heme 4-8 heme oxygenase 1 Homo sapiens 75-79 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Biliverdine 184-194 heme oxygenase 1 Homo sapiens 75-79 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Bilirubin 199-208 heme oxygenase 1 Homo sapiens 75-79 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Iron 262-266 heme oxygenase 1 Homo sapiens 75-79 16473885-4 2006 Similarly, long-term survival induced by DST was ablated when HO-1 activity was blocked by zinc protoporphyrin IX (ZnPPIX). zinc protoporphyrin 91-113 heme oxygenase 1 Homo sapiens 62-66 16473885-4 2006 Similarly, long-term survival induced by DST was ablated when HO-1 activity was blocked by zinc protoporphyrin IX (ZnPPIX). zinc protoporphyrin 115-121 heme oxygenase 1 Homo sapiens 62-66 16473885-7 2006 However, long-term survival and (dominant peripheral) tolerance were readily induced when DST was combined with induction of HO-1 expression by cobalt protoporphyrin IX (CoPPIX). cobaltiprotoporphyrin 144-168 heme oxygenase 1 Homo sapiens 125-129 16473885-7 2006 However, long-term survival and (dominant peripheral) tolerance were readily induced when DST was combined with induction of HO-1 expression by cobalt protoporphyrin IX (CoPPIX). cobaltiprotoporphyrin 170-176 heme oxygenase 1 Homo sapiens 125-129 16704082-3 2006 The expression of HO-1 in dental pulp was detected by means of SABC immunohistochemical technology. sabc 63-67 heme oxygenase 1 Homo sapiens 18-22 16601269-1 2006 The heme oxygenases, which consist of constitutive and inducible isozymes (HO-1, HO-2), catalyze the rate-limiting step in the metabolic conversion of heme to the bile pigments (i.e., biliverdin and bilirubin) and thus constitute a major intracellular source of iron and carbon monoxide (CO). Carbon Monoxide 288-290 heme oxygenase 1 Homo sapiens 75-79 16537525-5 2006 This induction of HO-1 occurred within clinical LNO(2) concentration ranges, far exceeded responses to equimolar amounts of linoleic acid and oxidized linoleic acid, and rivaled that induced by hemin. Linoleic Acid 124-137 heme oxygenase 1 Homo sapiens 18-22 16582079-3 2006 A (GT)(n) dinucleotide repeat in the human HO-1 promoter modulates HO-1 gene expression and shows length polymorphism, which is grouped into three classes: class S (<27 repeats), class M (> or = 27, <33 repeats), and class L (> or = 33 repeats) alleles. (n) dinucleotide 6-22 heme oxygenase 1 Homo sapiens 43-47 16582079-3 2006 A (GT)(n) dinucleotide repeat in the human HO-1 promoter modulates HO-1 gene expression and shows length polymorphism, which is grouped into three classes: class S (<27 repeats), class M (> or = 27, <33 repeats), and class L (> or = 33 repeats) alleles. (n) dinucleotide 6-22 heme oxygenase 1 Homo sapiens 67-71 16460683-4 2006 Curcumin increased HO-1 and glutamyl cysteine ligase modulator (GCLM) expression and stimulated Nrf2 binding to the ARE. Curcumin 0-8 heme oxygenase 1 Homo sapiens 19-23 16460683-5 2006 Curcumin also rapidly stimulated PKC phosphorylation and Ro-31-8220, a pan-PKC inhibitor, decreased curcumin-induced GCLM and HO-1 mRNA expression and ARE binding. Curcumin 0-8 heme oxygenase 1 Homo sapiens 126-130 16460683-5 2006 Curcumin also rapidly stimulated PKC phosphorylation and Ro-31-8220, a pan-PKC inhibitor, decreased curcumin-induced GCLM and HO-1 mRNA expression and ARE binding. Curcumin 100-108 heme oxygenase 1 Homo sapiens 126-130 16460683-6 2006 Rottlerin (a PKC delta inhibitor) and PKC delta antisense oligonucleotides significantly inhibited curcumin-induced GCLM and HO-1 mRNA expression and ARE binding. rottlerin 0-9 heme oxygenase 1 Homo sapiens 125-129 16460683-6 2006 Rottlerin (a PKC delta inhibitor) and PKC delta antisense oligonucleotides significantly inhibited curcumin-induced GCLM and HO-1 mRNA expression and ARE binding. Oligonucleotides 58-74 heme oxygenase 1 Homo sapiens 125-129 16460683-6 2006 Rottlerin (a PKC delta inhibitor) and PKC delta antisense oligonucleotides significantly inhibited curcumin-induced GCLM and HO-1 mRNA expression and ARE binding. Curcumin 99-107 heme oxygenase 1 Homo sapiens 125-129 16537525-0 2006 Fatty acid transduction of nitric oxide signaling: nitrolinoleic acid potently activates endothelial heme oxygenase 1 expression. Fatty Acids 0-10 heme oxygenase 1 Homo sapiens 101-117 16537525-0 2006 Fatty acid transduction of nitric oxide signaling: nitrolinoleic acid potently activates endothelial heme oxygenase 1 expression. Nitric Oxide 27-39 heme oxygenase 1 Homo sapiens 101-117 16537525-0 2006 Fatty acid transduction of nitric oxide signaling: nitrolinoleic acid potently activates endothelial heme oxygenase 1 expression. 10-nitro-9,12-octadecadienoic acid 51-69 heme oxygenase 1 Homo sapiens 101-117 16537525-5 2006 This induction of HO-1 occurred within clinical LNO(2) concentration ranges, far exceeded responses to equimolar amounts of linoleic acid and oxidized linoleic acid, and rivaled that induced by hemin. L-Leucyl-Hydroxylamine 48-51 heme oxygenase 1 Homo sapiens 18-22 16537525-5 2006 This induction of HO-1 occurred within clinical LNO(2) concentration ranges, far exceeded responses to equimolar amounts of linoleic acid and oxidized linoleic acid, and rivaled that induced by hemin. Linoleic Acid 151-164 heme oxygenase 1 Homo sapiens 18-22 16537525-7 2006 Actinomycin D inhibited LNO(2) induction of HO-1 in human aortic endothelial cells, and LNO(2) activated a 4.5-kb human HO-1 promoter construct, indicating transcriptional regulation of the HO-1 gene. Dactinomycin 0-13 heme oxygenase 1 Homo sapiens 44-48 16537525-7 2006 Actinomycin D inhibited LNO(2) induction of HO-1 in human aortic endothelial cells, and LNO(2) activated a 4.5-kb human HO-1 promoter construct, indicating transcriptional regulation of the HO-1 gene. L-Leucyl-Hydroxylamine 24-27 heme oxygenase 1 Homo sapiens 44-48 16537525-9 2006 The NO scavengers 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide and oxymyoglobin partially reversed induction of HO-1 by LNO(2), revealing that LNO(2) regulates HO-1 expression by predominantly NO-independent mechanisms. 1,3-dihydroxy-4,4,5,5-tetramethyl-2-(4-carboxyphenyl)tetrahydroimidazole 18-83 heme oxygenase 1 Homo sapiens 133-137 16537525-9 2006 The NO scavengers 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide and oxymyoglobin partially reversed induction of HO-1 by LNO(2), revealing that LNO(2) regulates HO-1 expression by predominantly NO-independent mechanisms. L-Leucyl-Hydroxylamine 141-144 heme oxygenase 1 Homo sapiens 133-137 16537525-9 2006 The NO scavengers 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide and oxymyoglobin partially reversed induction of HO-1 by LNO(2), revealing that LNO(2) regulates HO-1 expression by predominantly NO-independent mechanisms. L-Leucyl-Hydroxylamine 141-144 heme oxygenase 1 Homo sapiens 181-185 16537525-10 2006 In summary, the metabolic and inflammatory signaling actions of nitroalkenes can be transduced by robust HO-1 induction. nitroalkenes 64-76 heme oxygenase 1 Homo sapiens 105-109 16468956-5 2006 Therefore, we examined the impact of donor treatment with the selective inducer of HO-1, cobalt protoporphyrin (CoPP), on organ quality and transplant outcome in a standardized BD model in a F344-->LEW kidney transplant rat model. cobaltiprotoporphyrin 89-110 heme oxygenase 1 Homo sapiens 83-87 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Heme 115-119 heme oxygenase 1 Homo sapiens 0-16 16468956-5 2006 Therefore, we examined the impact of donor treatment with the selective inducer of HO-1, cobalt protoporphyrin (CoPP), on organ quality and transplant outcome in a standardized BD model in a F344-->LEW kidney transplant rat model. cobaltiprotoporphyrin 112-116 heme oxygenase 1 Homo sapiens 83-87 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Heme 115-119 heme oxygenase 1 Homo sapiens 18-22 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 0-16 16468956-6 2006 Immediately after BD induction, donor animals were administered a single dose of CoPP (5 mg/kg) and in control groups, HO-1 activity was blocked with zinc protoporphyrin (ZnPP, 20 mg/kg). zinc protoporphyrin 150-169 heme oxygenase 1 Homo sapiens 119-123 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 18-22 16468956-6 2006 Immediately after BD induction, donor animals were administered a single dose of CoPP (5 mg/kg) and in control groups, HO-1 activity was blocked with zinc protoporphyrin (ZnPP, 20 mg/kg). zinc protoporphyrin 171-175 heme oxygenase 1 Homo sapiens 119-123 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Biliverdine 170-172 heme oxygenase 1 Homo sapiens 0-16 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Biliverdine 170-172 heme oxygenase 1 Homo sapiens 18-22 16468956-8 2006 Blockade of HO-1 with ZnPP decreased the survival rates (p < 0.05) comparable to untreated brain-dead donors. zinc protoporphyrin 22-26 heme oxygenase 1 Homo sapiens 12-16 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Carbon Monoxide 175-190 heme oxygenase 1 Homo sapiens 0-16 16677090-5 2006 Of the various HSPs, heme oxygenase-I (HO-1), by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 84-99 heme oxygenase 1 Homo sapiens 39-43 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Carbon Monoxide 175-190 heme oxygenase 1 Homo sapiens 18-22 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Carbon Monoxide 192-194 heme oxygenase 1 Homo sapiens 0-16 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Carbon Monoxide 192-194 heme oxygenase 1 Homo sapiens 18-22 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Iron 201-205 heme oxygenase 1 Homo sapiens 0-16 16461755-1 2006 Heme oxygenase 1 (HO-1) is induced in response to cellular stress and is responsible for converting the prooxidant heme molecule into equimolar quantities of biliverdin (BV), carbon monoxide (CO), and iron. Iron 201-205 heme oxygenase 1 Homo sapiens 18-22 16385084-4 2006 Cortical 15d-PGJ2 in Adv-COX-1-treated rats was increased by 3-fold over control, which was correlated with reduced infarct volume and activated caspase 3, and increased peroxisome proliferator activated receptor-gamma (PPARgamma) and heme oxygenase-1 (HO-1). 15-deoxy-delta(12,14)-prostaglandin J2 9-17 heme oxygenase 1 Homo sapiens 253-257 16385084-7 2006 15d-PGJ2 and rosiglitazone at low concentrations suppressed H2O2-induced rat or human neuronal apoptosis and necrosis and induced PPARgamma and HO-1 expression. 15-deoxy-delta(12,14)-prostaglandin J2 0-8 heme oxygenase 1 Homo sapiens 144-148 16557439-4 2006 The isoform hemeoxygenase-1 is inducible by oxidative stress and may mediate cytoprotection mainly attributable to endogenously produced carbon monoxide. Carbon Monoxide 137-152 heme oxygenase 1 Homo sapiens 12-27 16385084-7 2006 15d-PGJ2 and rosiglitazone at low concentrations suppressed H2O2-induced rat or human neuronal apoptosis and necrosis and induced PPARgamma and HO-1 expression. Rosiglitazone 13-26 heme oxygenase 1 Homo sapiens 144-148 16677090-5 2006 Of the various HSPs, heme oxygenase-I (HO-1), by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 127-136 heme oxygenase 1 Homo sapiens 39-43 27277612-6 2006 When a potent antioxidant, N-acetyl-L-cysteine, was added to the culture medium before or after sonication, the induction was attenuated, indicating that reactive oxygen species are involved in HO-1 induction. Acetylcysteine 27-46 heme oxygenase 1 Homo sapiens 194-198 16222706-2 2006 Up-regulation of HO-1 in rat astroglia has been shown to facilitate iron sequestration by the mitochondrial compartment. Iron 68-72 heme oxygenase 1 Homo sapiens 17-21 16222706-9 2006 Glial HO-1 hyperactivity may contribute to cellular oxidative stress, pathological iron deposition, and bioenergetic failure characteristic of degenerating and inflamed neural tissues and may constitute a rational target for therapeutic intervention in these conditions. Iron 83-87 heme oxygenase 1 Homo sapiens 6-10 27277612-6 2006 When a potent antioxidant, N-acetyl-L-cysteine, was added to the culture medium before or after sonication, the induction was attenuated, indicating that reactive oxygen species are involved in HO-1 induction. Reactive Oxygen Species 154-177 heme oxygenase 1 Homo sapiens 194-198 16480975-0 2006 Heme oxygenase 1 mediates anti-inflammatory effects of 2",4",6"-tris(methoxymethoxy) chalcone. 2',4',6'-tris(methoxymethoxy) chalcone 55-93 heme oxygenase 1 Homo sapiens 0-16 16386335-10 2006 The synaptosomes isolated from gerbil pre-injected with FAEE and subsequently treated with AAPH or Fe(2+)/H(2)O(2) showed induction of heme oxygenase (HO-1) and heat shock protein 70 (HSP-70) but reduced inducible nitric oxide synthase (iNOS) levels. ethyl ferulate 56-60 heme oxygenase 1 Homo sapiens 151-155 16386335-10 2006 The synaptosomes isolated from gerbil pre-injected with FAEE and subsequently treated with AAPH or Fe(2+)/H(2)O(2) showed induction of heme oxygenase (HO-1) and heat shock protein 70 (HSP-70) but reduced inducible nitric oxide synthase (iNOS) levels. 2,2'-azobis(2-amidinopropane) 91-95 heme oxygenase 1 Homo sapiens 151-155 16386335-10 2006 The synaptosomes isolated from gerbil pre-injected with FAEE and subsequently treated with AAPH or Fe(2+)/H(2)O(2) showed induction of heme oxygenase (HO-1) and heat shock protein 70 (HSP-70) but reduced inducible nitric oxide synthase (iNOS) levels. Iron 99-101 heme oxygenase 1 Homo sapiens 151-155 16386335-10 2006 The synaptosomes isolated from gerbil pre-injected with FAEE and subsequently treated with AAPH or Fe(2+)/H(2)O(2) showed induction of heme oxygenase (HO-1) and heat shock protein 70 (HSP-70) but reduced inducible nitric oxide synthase (iNOS) levels. h(2)o 106-111 heme oxygenase 1 Homo sapiens 151-155 16498227-6 2006 We note the ability of Cd to elicit "oxidative stress" and to alter metal homeostasis notably of zinc which may lead to augmentation of the defense mechanisms involving induction of the antioxidant enzyme heme oxygenase-1 (HO-1) and the metal binding protein metallothionein (MT) in the kidney. Cadmium 23-25 heme oxygenase 1 Homo sapiens 205-221 16498227-6 2006 We note the ability of Cd to elicit "oxidative stress" and to alter metal homeostasis notably of zinc which may lead to augmentation of the defense mechanisms involving induction of the antioxidant enzyme heme oxygenase-1 (HO-1) and the metal binding protein metallothionein (MT) in the kidney. Cadmium 23-25 heme oxygenase 1 Homo sapiens 223-227 16498227-6 2006 We note the ability of Cd to elicit "oxidative stress" and to alter metal homeostasis notably of zinc which may lead to augmentation of the defense mechanisms involving induction of the antioxidant enzyme heme oxygenase-1 (HO-1) and the metal binding protein metallothionein (MT) in the kidney. Metals 68-73 heme oxygenase 1 Homo sapiens 205-221 16498227-6 2006 We note the ability of Cd to elicit "oxidative stress" and to alter metal homeostasis notably of zinc which may lead to augmentation of the defense mechanisms involving induction of the antioxidant enzyme heme oxygenase-1 (HO-1) and the metal binding protein metallothionein (MT) in the kidney. Metals 68-73 heme oxygenase 1 Homo sapiens 223-227 16498227-7 2006 We hypothesize that renal Cd accumulation triggers the host responses mediated by HO-1 and MT in an attempt to protect the kidney against injurious oxidative stress and to resist a rise in blood pressure levels. Cadmium 26-28 heme oxygenase 1 Homo sapiens 82-93 16498227-8 2006 This hypothesis predicts that individuals with less active HO-1 (caused by the HO-1 genetic polymorphisms) are more likely to have renal injury and express a hypertensive phenotype following chronic ingestion of low-level Cd, compared with those having more active HO-1. Cadmium 222-224 heme oxygenase 1 Homo sapiens 59-63 16498227-8 2006 This hypothesis predicts that individuals with less active HO-1 (caused by the HO-1 genetic polymorphisms) are more likely to have renal injury and express a hypertensive phenotype following chronic ingestion of low-level Cd, compared with those having more active HO-1. Cadmium 222-224 heme oxygenase 1 Homo sapiens 79-83 16498227-8 2006 This hypothesis predicts that individuals with less active HO-1 (caused by the HO-1 genetic polymorphisms) are more likely to have renal injury and express a hypertensive phenotype following chronic ingestion of low-level Cd, compared with those having more active HO-1. Cadmium 222-224 heme oxygenase 1 Homo sapiens 79-83 16495813-5 2006 Pretreatment with curcumin protected hepatocytes in a model of oxidative injury and this protection was mediated through HO-1. Curcumin 18-26 heme oxygenase 1 Homo sapiens 121-125 16495813-6 2006 In a model of cold preservation injury, curcumin pretreatment resulted in elevation of HO-1 throughout the cold storage and rewarming period, and was cytoprotective against oxidative injury. Curcumin 40-48 heme oxygenase 1 Homo sapiens 87-91 16495813-7 2006 This is the first study to demonstrate that curcumin induces HO-1 in human hepatocytes, and that the protective effects of curcumin pretreatment may have clinical potential in hepatic transplantation. Curcumin 44-52 heme oxygenase 1 Homo sapiens 61-65 16480975-2 2006 We also demonstrate that TMMC by itself is a potent inducer of heme oxygenase 1 (HO-1). 2',4',6'-tris(methoxymethoxy) chalcone 25-29 heme oxygenase 1 Homo sapiens 63-79 16480975-2 2006 We also demonstrate that TMMC by itself is a potent inducer of heme oxygenase 1 (HO-1). 2',4',6'-tris(methoxymethoxy) chalcone 25-29 heme oxygenase 1 Homo sapiens 81-85 16480975-4 2006 Treating cells with the specific p42/44 MAPK inhibitor, PD98059, blocked the TMMC-mediated induction of HO-1 and the inhibition of LPS-stimulated expression of iNOS. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 56-63 heme oxygenase 1 Homo sapiens 104-108 16480975-4 2006 Treating cells with the specific p42/44 MAPK inhibitor, PD98059, blocked the TMMC-mediated induction of HO-1 and the inhibition of LPS-stimulated expression of iNOS. 2',4',6'-tris(methoxymethoxy) chalcone 77-81 heme oxygenase 1 Homo sapiens 104-108 16480975-6 2006 Our data suggest that TMMC exerts an anti-inflammatory effect in macrophages through a mechanism that involves the induction of HO-1, which is mediated by activation of p42/44 MAPK and GSH depletion. 2',4',6'-tris(methoxymethoxy) chalcone 22-26 heme oxygenase 1 Homo sapiens 128-132 16480975-6 2006 Our data suggest that TMMC exerts an anti-inflammatory effect in macrophages through a mechanism that involves the induction of HO-1, which is mediated by activation of p42/44 MAPK and GSH depletion. Glutathione 185-188 heme oxygenase 1 Homo sapiens 128-132 16495813-4 2006 This study investigates in detail the effect of curcumin on the stress-response in human hepatocytes, in particular its effect on heme oxygenase 1 (HO-1) and its cytoprotective effect. Curcumin 48-56 heme oxygenase 1 Homo sapiens 130-146 16495813-4 2006 This study investigates in detail the effect of curcumin on the stress-response in human hepatocytes, in particular its effect on heme oxygenase 1 (HO-1) and its cytoprotective effect. Curcumin 48-56 heme oxygenase 1 Homo sapiens 148-152 16337498-4 2006 RESULTS: HO-1 as well as ecNOS gene and protein expression significantly increased upon Carnitines incubation. Carnitine 88-98 heme oxygenase 1 Homo sapiens 9-13 16337498-7 2006 Coincubation of C (0.5-1.0-2.0 mM), AC (0.1-0.2-0.4 mM) and PC (0.05-0.1-0.2 mM) with H2O2 further increased HO-1 gene expression and not only normalized vs. H2O2 but even increased vs. basal ecNOS mRNA. Hydrogen Peroxide 86-90 heme oxygenase 1 Homo sapiens 109-113 16337498-9 2006 CONCLUSION: This is the first report that has utilized a molecular biological approach to demonstrate a direct stimulatory effect of Carnitines on gene and protein expression of the oxidative stress related markers HO-1 and ecNOS. Carnitine 133-143 heme oxygenase 1 Homo sapiens 215-219 16337498-10 2006 As HO-1 and NO are known as antioxidant, antiproliferative and anti-inflammatory, their increased expression would be expected to protect from oxidative stress related cardiovascular risk factors and myocardial damage, therefore adding this effect to the multiple pathways involved in the effects of carnitines. Carnitine 300-310 heme oxygenase 1 Homo sapiens 3-7 16329999-0 2006 CO from enhanced HO activity or from CORM-2 inhibits both O2- and NO production and downregulates HO-1 expression in LPS-stimulated macrophages. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 98-102 16243536-0 2006 Piceatannol upregulates endothelial heme oxygenase-1 expression via novel protein kinase C and tyrosine kinase pathways. 3,3',4,5'-tetrahydroxystilbene 0-11 heme oxygenase 1 Homo sapiens 36-52 16243536-3 2006 In this study, we examined the ability of piceatannol to upregulate HO-1 expression in endothelial cells. 3,3',4,5'-tetrahydroxystilbene 42-53 heme oxygenase 1 Homo sapiens 68-72 16243536-4 2006 We found piceatannol at micromolar (10-50 microM) concentrations dramatically increased HO-1 protein levels in a time-dependent manner. 3,3',4,5'-tetrahydroxystilbene 9-20 heme oxygenase 1 Homo sapiens 88-92 16243536-5 2006 Piceatannol was similarly potent in the induction of HO-1 as hemin, arsenate, and 15d-PGJ2, and was more potent than some other phytochemicals including curcumin, EGCG, baicalein, and quercetin. 3,3',4,5'-tetrahydroxystilbene 0-11 heme oxygenase 1 Homo sapiens 53-57 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 92-95 heme oxygenase 1 Homo sapiens 214-218 16482620-6 2006 CONCLUSION: PGG up-regulates HO-1 expression by stimulating Nrf2 nuclear translocation in an ERK-dependent manner, and HO-1 expression by PGG may serve as one of the important mechanisms for its hepatoprotective effects. beta-penta-O-galloyl-glucose 12-15 heme oxygenase 1 Homo sapiens 29-33 16482620-6 2006 CONCLUSION: PGG up-regulates HO-1 expression by stimulating Nrf2 nuclear translocation in an ERK-dependent manner, and HO-1 expression by PGG may serve as one of the important mechanisms for its hepatoprotective effects. beta-penta-O-galloyl-glucose 138-141 heme oxygenase 1 Homo sapiens 119-123 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 117-121 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Heme 34-38 heme oxygenase 1 Homo sapiens 99-115 16254033-5 2006 Donor treatment with bilirubin up-regulated mRNA expression of protective genes such as HO-1 and bcl-2 and suppressed proinflammatory and proapoptotic genes including monocyte chemoattractant protein-1 and caspase-3 and -8 in the islet grafts before transplantation. Bilirubin 21-30 heme oxygenase 1 Homo sapiens 88-92 16137675-3 2006 Biodegradation of extravasated hemoglobin (exvHb) and deposition of iron in alveoli occurred at 3-56 h post-exposure and was preceded by LKC degranulation and accumulation of MPO, HO-1, and SOD-1 in HLs. Iron 68-72 heme oxygenase 1 Homo sapiens 180-184 16474202-4 2006 Increased HO-1 induction by hemin resulted in a significant decrease in the Lyso-PC-mediated induction of MCP-1 mRNA expression. Hemin 28-33 heme oxygenase 1 Homo sapiens 10-14 16474202-4 2006 Increased HO-1 induction by hemin resulted in a significant decrease in the Lyso-PC-mediated induction of MCP-1 mRNA expression. Lysophosphatidylcholines 76-83 heme oxygenase 1 Homo sapiens 10-14 16474202-5 2006 SnPP (IX), the specific inhibitor of HO-1 enzymatic activity, prevented the hemin-mediated attenuation of MCP-1 mRNA expression. S-Nitroso-N-propionyl-D,L-penicillamine 0-4 heme oxygenase 1 Homo sapiens 37-41 16474202-5 2006 SnPP (IX), the specific inhibitor of HO-1 enzymatic activity, prevented the hemin-mediated attenuation of MCP-1 mRNA expression. Hemin 76-81 heme oxygenase 1 Homo sapiens 37-41 16243536-8 2006 Piceatannol-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine and glutathione, but not by other antioxidants. 3,3',4,5'-tetrahydroxystilbene 0-11 heme oxygenase 1 Homo sapiens 21-25 16243536-8 2006 Piceatannol-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine and glutathione, but not by other antioxidants. Acetylcysteine 69-85 heme oxygenase 1 Homo sapiens 21-25 16243536-8 2006 Piceatannol-mediated HO-1 induction was abrogated in the presence of N-acetylcysteine and glutathione, but not by other antioxidants. Glutathione 90-101 heme oxygenase 1 Homo sapiens 21-25 16243536-9 2006 Induction of HO-1 by piceatannol was further enhanced by using buthionine sulfoximine. 3,3',4,5'-tetrahydroxystilbene 21-32 heme oxygenase 1 Homo sapiens 13-17 16243536-9 2006 Induction of HO-1 by piceatannol was further enhanced by using buthionine sulfoximine. Buthionine Sulfoximine 63-85 heme oxygenase 1 Homo sapiens 13-17 16243536-10 2006 In addition, we determined that tyrosine kinase was involved in the induction of HO-1 by using tyrosine kinase inhibitors, herbimycin A, erbstatin, and genistein; in contrast, no significant changes in the pretreatment of PI3 kinase or MAP kinase inhibitors was determined. herbimycin 123-135 heme oxygenase 1 Homo sapiens 81-85 16243536-10 2006 In addition, we determined that tyrosine kinase was involved in the induction of HO-1 by using tyrosine kinase inhibitors, herbimycin A, erbstatin, and genistein; in contrast, no significant changes in the pretreatment of PI3 kinase or MAP kinase inhibitors was determined. erbstatin 137-146 heme oxygenase 1 Homo sapiens 81-85 16243536-10 2006 In addition, we determined that tyrosine kinase was involved in the induction of HO-1 by using tyrosine kinase inhibitors, herbimycin A, erbstatin, and genistein; in contrast, no significant changes in the pretreatment of PI3 kinase or MAP kinase inhibitors was determined. Genistein 152-161 heme oxygenase 1 Homo sapiens 81-85 16243536-11 2006 HO-1 induction was blocked by the protein kinase C inhibitors calphostin C, rottlerin, and long PMA pretreatment, whereas conventional PKC inhibitors, Go6976, and Ca2+ chelator BAPTA/AM, had no effect. rottlerin 76-85 heme oxygenase 1 Homo sapiens 0-4 16243536-11 2006 HO-1 induction was blocked by the protein kinase C inhibitors calphostin C, rottlerin, and long PMA pretreatment, whereas conventional PKC inhibitors, Go6976, and Ca2+ chelator BAPTA/AM, had no effect. Tetradecanoylphorbol Acetate 96-99 heme oxygenase 1 Homo sapiens 0-4 16243536-11 2006 HO-1 induction was blocked by the protein kinase C inhibitors calphostin C, rottlerin, and long PMA pretreatment, whereas conventional PKC inhibitors, Go6976, and Ca2+ chelator BAPTA/AM, had no effect. Go 6976 151-157 heme oxygenase 1 Homo sapiens 0-4 16243536-11 2006 HO-1 induction was blocked by the protein kinase C inhibitors calphostin C, rottlerin, and long PMA pretreatment, whereas conventional PKC inhibitors, Go6976, and Ca2+ chelator BAPTA/AM, had no effect. 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid 177-182 heme oxygenase 1 Homo sapiens 0-4 16243536-13 2006 Treating ECs with zinc protoporphyrin, an HO-1 inhibito blocked the anti-inflammatory effect of piceatannol. zinc protoporphyrin 18-37 heme oxygenase 1 Homo sapiens 42-46 16243536-13 2006 Treating ECs with zinc protoporphyrin, an HO-1 inhibito blocked the anti-inflammatory effect of piceatannol. 3,3',4,5'-tetrahydroxystilbene 96-107 heme oxygenase 1 Homo sapiens 42-46 16243536-14 2006 In summary, this study identified piceatannol as a novel phytochemical inducer of HO-1 expression and identified the mechanisms involved in this process. 3,3',4,5'-tetrahydroxystilbene 34-45 heme oxygenase 1 Homo sapiens 82-86 16233958-4 2006 In the present study, we demonstrate that systemically administered SUL can enter the brain as determined by increased mRNA and protein levels of the Nrf2-responsive gene heme oxygenase 1 (HO-1). sulforaphane 68-71 heme oxygenase 1 Homo sapiens 171-187 16482620-0 2006 1,2,3,4,6-penta-O-galloyl-beta-D-glucose up-regulates heme oxygenase-1 expression by stimulating Nrf2 nuclear translocation in an extracellular signal-regulated kinase-dependent manner in HepG2 cells. beta-penta-O-galloyl-glucose 0-40 heme oxygenase 1 Homo sapiens 54-70 16482620-1 2006 AIM: To examine the potency of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG) as a hepatic heme oxygenase-1 (HO-1) inducer and its regulation in HepG2 cells. beta-penta-O-galloyl-glucose 31-71 heme oxygenase 1 Homo sapiens 91-107 16482620-1 2006 AIM: To examine the potency of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG) as a hepatic heme oxygenase-1 (HO-1) inducer and its regulation in HepG2 cells. beta-penta-O-galloyl-glucose 31-71 heme oxygenase 1 Homo sapiens 109-113 16482620-1 2006 AIM: To examine the potency of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG) as a hepatic heme oxygenase-1 (HO-1) inducer and its regulation in HepG2 cells. beta-penta-O-galloyl-glucose 73-76 heme oxygenase 1 Homo sapiens 91-107 16482620-1 2006 AIM: To examine the potency of 1,2,3,4,6-penta-O-galloyl-beta-D-glucose (PGG) as a hepatic heme oxygenase-1 (HO-1) inducer and its regulation in HepG2 cells. beta-penta-O-galloyl-glucose 73-76 heme oxygenase 1 Homo sapiens 109-113 16482620-4 2006 RESULTS: PGG up-regulated HO-1 expression and this expression conferred cytoprotection against oxidative injury induced by t-butyl hydroperoxide. beta-penta-O-galloyl-glucose 9-12 heme oxygenase 1 Homo sapiens 26-30 16482620-4 2006 RESULTS: PGG up-regulated HO-1 expression and this expression conferred cytoprotection against oxidative injury induced by t-butyl hydroperoxide. tert-Butylhydroperoxide 123-144 heme oxygenase 1 Homo sapiens 26-30 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 10-13 heme oxygenase 1 Homo sapiens 104-108 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 10-13 heme oxygenase 1 Homo sapiens 214-218 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 92-95 heme oxygenase 1 Homo sapiens 104-108 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 92-95 heme oxygenase 1 Homo sapiens 214-218 16482620-5 2006 Moreover, PGG induced Nrf2 nuclear translocation, which was found to be an upstream step of PGG-induced HO-1 expression, and ERK activation, of which pathway was involved in PGG-induced Nrf2 nuclear translocation, HO-1 expression and cytoprotection. beta-penta-O-galloyl-glucose 92-95 heme oxygenase 1 Homo sapiens 104-108 16388581-2 2006 The heme orientation within the active site, which is thought to determine the oxidation regiospecificity, is shown here for the human enzyme (hHO1) to be largely determined by interactions between the heme carboxylic acid groups and residues Arg183 and Lys18 but not Tyr134. Heme 4-8 heme oxygenase 1 Homo sapiens 143-147 16388581-2 2006 The heme orientation within the active site, which is thought to determine the oxidation regiospecificity, is shown here for the human enzyme (hHO1) to be largely determined by interactions between the heme carboxylic acid groups and residues Arg183 and Lys18 but not Tyr134. heme carboxylic acid 202-222 heme oxygenase 1 Homo sapiens 143-147 16388581-2 2006 The heme orientation within the active site, which is thought to determine the oxidation regiospecificity, is shown here for the human enzyme (hHO1) to be largely determined by interactions between the heme carboxylic acid groups and residues Arg183 and Lys18 but not Tyr134. octadeca(lysine) 254-259 heme oxygenase 1 Homo sapiens 143-147 16388581-6 2006 Kinetic studies demonstrate that mutations of Arg183 greatly impair the rate of the P450 reductase-dependent reaction, in accord with the earlier finding that Arg183 is involved in binding of the reductase to hHO1, but have little effect on the ascorbate reaction. Ascorbic Acid 245-254 heme oxygenase 1 Homo sapiens 209-213 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Heme 34-38 heme oxygenase 1 Homo sapiens 117-121 16329999-11 2006 However, endogenous overproduction of CO by super-induction of HO-1 (obtained by pretreatment of macrophages with either buthionine sulfoximine or hemin) decreased the LPS-derived iNOS expression without affecting cell survival. Carbon Monoxide 38-40 heme oxygenase 1 Homo sapiens 63-67 16329999-11 2006 However, endogenous overproduction of CO by super-induction of HO-1 (obtained by pretreatment of macrophages with either buthionine sulfoximine or hemin) decreased the LPS-derived iNOS expression without affecting cell survival. Buthionine Sulfoximine 121-143 heme oxygenase 1 Homo sapiens 63-67 16329999-11 2006 However, endogenous overproduction of CO by super-induction of HO-1 (obtained by pretreatment of macrophages with either buthionine sulfoximine or hemin) decreased the LPS-derived iNOS expression without affecting cell survival. Hemin 147-152 heme oxygenase 1 Homo sapiens 63-67 16329999-13 2006 Thus, upregulation of HO-1 and overproduction of CO may allow the survival of LPS-stimulated macrophages; first, by eliminating the free heme to prevent Fenton reaction, second, by limiting the availability of free heme required for induction of NO-producing heme enzyme (i.e., iNOS), third, by limiting additional production of O(2)(-) and NO via CO-derived inhibition on the activities of heme enzymes like NADPH oxidase and iNOS, respectively. Heme 137-141 heme oxygenase 1 Homo sapiens 22-26 16329999-13 2006 Thus, upregulation of HO-1 and overproduction of CO may allow the survival of LPS-stimulated macrophages; first, by eliminating the free heme to prevent Fenton reaction, second, by limiting the availability of free heme required for induction of NO-producing heme enzyme (i.e., iNOS), third, by limiting additional production of O(2)(-) and NO via CO-derived inhibition on the activities of heme enzymes like NADPH oxidase and iNOS, respectively. Heme 215-219 heme oxygenase 1 Homo sapiens 22-26 16329999-13 2006 Thus, upregulation of HO-1 and overproduction of CO may allow the survival of LPS-stimulated macrophages; first, by eliminating the free heme to prevent Fenton reaction, second, by limiting the availability of free heme required for induction of NO-producing heme enzyme (i.e., iNOS), third, by limiting additional production of O(2)(-) and NO via CO-derived inhibition on the activities of heme enzymes like NADPH oxidase and iNOS, respectively. Superoxides 329-333 heme oxygenase 1 Homo sapiens 22-26 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 99-115 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 117-121 16329999-1 2006 Carbon monoxide (CO) arising from heme degradation, catalyzed particularly by the stress-inducible heme oxygenase-1 (HO-1), has recently been demonstrated to provide cytoprotection against cell death in macrophages stimulated with bacterial lipopolysaccharide (LPS). Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 99-115 16629181-7 2006 These results suggested that heme oxygenase-1 mediates heme iron influx and efflux in intestinal cells. Iron 60-64 heme oxygenase 1 Homo sapiens 29-45 17302375-5 2006 Enhancing cellular thiols with N-acetylcystein prevented the NFV-induced drop in reduced thiols and partially protected against the induction in HO-1, but failed to prevent insulin resistance or cleavage of poly ADP ribose polymerase (PARP), a process indicative of activation of pro-apoptotic caspases. Sulfhydryl Compounds 19-25 heme oxygenase 1 Homo sapiens 145-149 16629181-0 2006 Heme oxygenase 1 overexpression increases iron fluxes in caco-2 cells. Iron 42-46 heme oxygenase 1 Homo sapiens 0-16 16487037-8 2006 Expression of the enzyme heme oxygenase-1 (HO-1), a known Nrf2 target gene, was upregulated by exposure of JAR cells to arsenic. Arsenic 120-127 heme oxygenase 1 Homo sapiens 25-41 16633995-5 2006 EPO cellular mechanisms are not completely known, and the identification of close biochemical and molecular relationships between EPO and heme oxygenase-1 (HO-1), which has potent antioxidant and anti-apoptotic properties, could provide the rationale for the beneficial effect of carnitine having been shown to possess antioxidant, anti-apoptotic and erythropoietic activities and to induce HO-1 expression, not only in dialysis patients who fail to respond adequately to EPO, but also in patients with heart failure. Carnitine 280-289 heme oxygenase 1 Homo sapiens 138-154 16629181-2 2006 Heme oxygenase-1 participates in the cleavage of the heme ring producing biliverdin, CO and ferrous Fe. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 16629181-2 2006 Heme oxygenase-1 participates in the cleavage of the heme ring producing biliverdin, CO and ferrous Fe. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 0-16 16629181-2 2006 Heme oxygenase-1 participates in the cleavage of the heme ring producing biliverdin, CO and ferrous Fe. Carbon Monoxide 85-87 heme oxygenase 1 Homo sapiens 0-16 16629181-2 2006 Heme oxygenase-1 participates in the cleavage of the heme ring producing biliverdin, CO and ferrous Fe. Iron 100-102 heme oxygenase 1 Homo sapiens 0-16 16943657-1 2006 Heme oxygenase 1 (HO-1) is an enzyme important in the catabolism of heme that is induced under conditions of oxidative stress. Heme 68-72 heme oxygenase 1 Homo sapiens 0-16 16943657-1 2006 Heme oxygenase 1 (HO-1) is an enzyme important in the catabolism of heme that is induced under conditions of oxidative stress. Heme 68-72 heme oxygenase 1 Homo sapiens 18-22 16943657-2 2006 HO-1 degradation of heme yields biliverdin, bilirubin, carbon monoxide and iron. Biliverdine 32-42 heme oxygenase 1 Homo sapiens 0-4 16943657-2 2006 HO-1 degradation of heme yields biliverdin, bilirubin, carbon monoxide and iron. Bilirubin 44-53 heme oxygenase 1 Homo sapiens 0-4 16943657-2 2006 HO-1 degradation of heme yields biliverdin, bilirubin, carbon monoxide and iron. Carbon Monoxide 55-70 heme oxygenase 1 Homo sapiens 0-4 16943657-2 2006 HO-1 degradation of heme yields biliverdin, bilirubin, carbon monoxide and iron. Iron 75-79 heme oxygenase 1 Homo sapiens 0-4 16633995-5 2006 EPO cellular mechanisms are not completely known, and the identification of close biochemical and molecular relationships between EPO and heme oxygenase-1 (HO-1), which has potent antioxidant and anti-apoptotic properties, could provide the rationale for the beneficial effect of carnitine having been shown to possess antioxidant, anti-apoptotic and erythropoietic activities and to induce HO-1 expression, not only in dialysis patients who fail to respond adequately to EPO, but also in patients with heart failure. Carnitine 280-289 heme oxygenase 1 Homo sapiens 156-160 16337271-1 2006 Solution proton NMR has been used here to show that, as either the high-spin ferric, protohemin (PH) substrate complex at neutral pH, or the low-spin ferric, cyanide-inhibited PH substrate complex, the active site electronic and molecular structure of the 233- and 265-residue recombinant constructs of human heme oxygenase-1, hHO, are essentially indistinguishable. Cyanides 158-165 heme oxygenase 1 Homo sapiens 309-325 16697692-5 2006 Heme oxygenase-1 induction by treatment with cobalt protoporphyrin IX resulted in resistance to apoptosis, activation of Akt, reduction in p21(Cip/WAF1) levels and modification of bcl2/bax ratio towards survival. cobaltiprotoporphyrin 45-69 heme oxygenase 1 Homo sapiens 0-16 16697692-8 2006 Protection against apoptosis in cells treated with cobalt protoporphyrin IX was reverted by incubation with heme oxygenase-1 small interfering RNA. cobaltiprotoporphyrin 51-75 heme oxygenase 1 Homo sapiens 108-124 16697692-9 2006 This study shows an antiapoptotic effect of heme oxygenase-1 in colon cancer cells which could be mediated by the formation of bilirubin and biliverdin. Bilirubin 127-136 heme oxygenase 1 Homo sapiens 44-60 16697692-9 2006 This study shows an antiapoptotic effect of heme oxygenase-1 in colon cancer cells which could be mediated by the formation of bilirubin and biliverdin. Biliverdine 141-151 heme oxygenase 1 Homo sapiens 44-60 16600087-0 2006 [Heme oxygenase-1 expression and apoptosis induced by cadmium in human embryon kidney cells]. Cadmium 54-61 heme oxygenase 1 Homo sapiens 1-17 16288501-7 2006 In particular, we examined the effect of anthocyanidins on endothelial heme oxygenase-1 (HO-1), an inducible stress protein. Anthocyanins 41-55 heme oxygenase 1 Homo sapiens 71-87 16401919-8 2006 Doxazosin concentration dependently reduced HO-1 gene and protein expression (0.57 +/- 0.07 vs. 0.49 +/- 0.06 d.u. Doxazosin 0-9 heme oxygenase 1 Homo sapiens 44-48 16401919-13 2006 Surprisingly, HO-1, which is induced and protected by oxidative stress, is also reduced by doxazosin. Doxazosin 91-100 heme oxygenase 1 Homo sapiens 14-18 16401919-15 2006 From the results of this preliminary study it could be proposed that the proapoptotic effect of doxazosin could be mediated, at least in part, through the contemporary inhibition of HO-1. Doxazosin 96-105 heme oxygenase 1 Homo sapiens 182-186 18958680-7 2006 Indirubin-3"-monoxime treatment also enhanced cyclo-oxygenase 2 expression in PMA-differentiated cells, but reduced the expression of macrophage inflammatory protein, catalase, heme oxygenase-1, glutathione peroxidase, and manganese superoxide dismutase; a scenario consistent with oxidative stress. indirubin-3'-monoxime 0-21 heme oxygenase 1 Homo sapiens 177-193 16374435-10 2006 Intradialytic removal of L-ascorbic acid, uric acid, bilirubin, 3-indoxyl sulfate, indoxyl-beta-D-glucuronide, p-cresol, and phenol increases the risk of LDL oxidation and subsequent endothelial cell damage, which underlines the importance of activation of cytoprotective HO-1 and ferritin in endothelium. Phenol 125-131 heme oxygenase 1 Homo sapiens 272-276 16374439-1 2006 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, producing carbon monoxide (CO), which carries potent antiproliferative and anti-inflammatory effects in the vascular walls. Carbon Monoxide 81-96 heme oxygenase 1 Homo sapiens 0-16 16374439-1 2006 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, producing carbon monoxide (CO), which carries potent antiproliferative and anti-inflammatory effects in the vascular walls. Carbon Monoxide 81-96 heme oxygenase 1 Homo sapiens 18-22 16374439-1 2006 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, producing carbon monoxide (CO), which carries potent antiproliferative and anti-inflammatory effects in the vascular walls. Carbon Monoxide 98-100 heme oxygenase 1 Homo sapiens 0-16 16374439-1 2006 Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, producing carbon monoxide (CO), which carries potent antiproliferative and anti-inflammatory effects in the vascular walls. Carbon Monoxide 98-100 heme oxygenase 1 Homo sapiens 18-22 16600087-1 2006 OBJECTIVE: To investigate apoptosis and expression of heme oxygenase-1 (HO-1) induced by cadmium in human embryonic kidney cells (HEK239T). Cadmium 89-96 heme oxygenase 1 Homo sapiens 54-70 16600087-1 2006 OBJECTIVE: To investigate apoptosis and expression of heme oxygenase-1 (HO-1) induced by cadmium in human embryonic kidney cells (HEK239T). Cadmium 89-96 heme oxygenase 1 Homo sapiens 72-76 16600087-6 2006 The CdCl(2) of between 10 and 40 micromol/L could highly induce the expression of HO-1 of the human embryonic kidney cells. cdcl 4-8 heme oxygenase 1 Homo sapiens 82-86 16600087-8 2006 CONCLUSION: The cadmium can induce the apoptosis of the human embryonic kidney cells and up-regulate the expression of HO-1. Cadmium 16-23 heme oxygenase 1 Homo sapiens 119-123 16183036-1 2005 In the beginning, the microsomal HO system was presumed to be made of one isozymes, now known as HO-1, which was cytP450-dependent; and, was thought to be of physiological significance solely in the context of catalysis of hemoglobin heme to bile pigments and CO. A succession of discoveries including characterization of the system as an independent mono-oxygenase, identification of a second form, called HO-2, free radical quenching activity of bile pigments, analogous function of CO in cell signaling to NO, and characterization of the system as HSP32 cognates has led to such an impressive expansion in the number of reports dealing with the HO system that surpass anyone"s expectation. Heme 234-238 heme oxygenase 1 Homo sapiens 97-101 16298686-4 2005 In neuron-glia cultures, we found that H2O2, a product of dopamine metabolism, or l-3,4-dihydroxyphenylalanine (L-DOPA), the dopamine precursor used in the therapy of PD, induced a fast up-regulation of HO-1 mRNA and protein levels, followed by a secondary down-regulation. Hydrogen Peroxide 39-43 heme oxygenase 1 Homo sapiens 203-207 16298686-4 2005 In neuron-glia cultures, we found that H2O2, a product of dopamine metabolism, or l-3,4-dihydroxyphenylalanine (L-DOPA), the dopamine precursor used in the therapy of PD, induced a fast up-regulation of HO-1 mRNA and protein levels, followed by a secondary down-regulation. Levodopa 82-110 heme oxygenase 1 Homo sapiens 203-207 16298686-4 2005 In neuron-glia cultures, we found that H2O2, a product of dopamine metabolism, or l-3,4-dihydroxyphenylalanine (L-DOPA), the dopamine precursor used in the therapy of PD, induced a fast up-regulation of HO-1 mRNA and protein levels, followed by a secondary down-regulation. Levodopa 112-118 heme oxygenase 1 Homo sapiens 203-207 16298686-5 2005 H2O2 and L-DOPA also increased HO-1 expression in astrocyte cultures, but with a delayed time course in H2O2-treated cultures. Hydrogen Peroxide 0-4 heme oxygenase 1 Homo sapiens 31-35 16298686-5 2005 H2O2 and L-DOPA also increased HO-1 expression in astrocyte cultures, but with a delayed time course in H2O2-treated cultures. Levodopa 9-15 heme oxygenase 1 Homo sapiens 31-35 16298686-7 2005 Because exogenously applied GDNF prevented HO-1 up-regulation in cultures treated with H2O2 or l-DOPA, and antibody neutralization of GDNF prevented the secondary HO-1 down-regulation observed in neuron-glia cultures, we propose that GDNF negatively modulates HO-1 expression induced by oxidative stress. Hydrogen Peroxide 87-91 heme oxygenase 1 Homo sapiens 43-47 16298686-7 2005 Because exogenously applied GDNF prevented HO-1 up-regulation in cultures treated with H2O2 or l-DOPA, and antibody neutralization of GDNF prevented the secondary HO-1 down-regulation observed in neuron-glia cultures, we propose that GDNF negatively modulates HO-1 expression induced by oxidative stress. Levodopa 95-101 heme oxygenase 1 Homo sapiens 43-47 16154530-7 2005 Ortiz de Montellano, Reaction intermediates and single turnover rate constants for the oxidation of heme by human heme oxygenase-1, J. Biol. Heme 100-104 heme oxygenase 1 Homo sapiens 114-130 16198234-2 2005 Heme oxygenase-1 (HO-1), implicated in a role in NO resistance, might confer its protective effect through the direct products biliverdin and CO or the secondary product bilirubin. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 0-16 16382109-3 2005 Inducible HO (HO-1) and constitutive HO (HO-2) are mostly recognized for their roles in the oxidation of heme and production of CO and biliverdin, whereas the biological function of the third HO isoform, HO-3, is still unclear. Heme 105-109 heme oxygenase 1 Homo sapiens 14-18 16382109-3 2005 Inducible HO (HO-1) and constitutive HO (HO-2) are mostly recognized for their roles in the oxidation of heme and production of CO and biliverdin, whereas the biological function of the third HO isoform, HO-3, is still unclear. Carbon Monoxide 128-130 heme oxygenase 1 Homo sapiens 14-18 16382109-3 2005 Inducible HO (HO-1) and constitutive HO (HO-2) are mostly recognized for their roles in the oxidation of heme and production of CO and biliverdin, whereas the biological function of the third HO isoform, HO-3, is still unclear. Biliverdine 135-145 heme oxygenase 1 Homo sapiens 14-18 16356130-1 2005 Heme oxygenase (HO)-1, involved in the heme degradation process, is an important antioxidant enzyme. Heme 39-43 heme oxygenase 1 Homo sapiens 0-21 16323392-4 2005 Other mediators such as endothelin-1 and the heme oxygenase-1/carbon monoxide system have recently been found to be important contributors. Carbon Monoxide 62-77 heme oxygenase 1 Homo sapiens 45-61 16198371-4 2005 Human coronary arteriolar endothelial cells exposed to resveratrol or Trx-1 on Matrigel demonstrated significantly accelerated tubular morphogenesis with induction of HO-1 and VEGF expression. Resveratrol 55-66 heme oxygenase 1 Homo sapiens 167-171 16208635-8 2005 Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. Heme 22-26 heme oxygenase 1 Homo sapiens 0-16 16208635-8 2005 Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. Porphyrins 59-68 heme oxygenase 1 Homo sapiens 0-16 16208635-8 2005 Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 0-16 16208635-8 2005 Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. Carbon Monoxide 97-112 heme oxygenase 1 Homo sapiens 0-16 16208635-8 2005 Heme oxygenase-1 is a heme-degrading enzyme that opens the porphyrin ring, producing biliverdin, carbon monoxide, and the most dangerous product - free redox active iron. Iron 165-169 heme oxygenase 1 Homo sapiens 0-16 16154535-0 2005 Heme oxygenase-1-derived carbon monoxide stimulates adenosine triphosphate generation in human hepatocyte. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 16154535-0 2005 Heme oxygenase-1-derived carbon monoxide stimulates adenosine triphosphate generation in human hepatocyte. Adenosine Triphosphate 52-74 heme oxygenase 1 Homo sapiens 0-16 16373841-0 2005 Mechanisms of heme oxygenase-1-mediated cardiac and pulmonary vascular protection in chronic hypoxia: roles of carbon monoxide and bilirubin. Carbon Monoxide 111-126 heme oxygenase 1 Homo sapiens 14-30 16373841-0 2005 Mechanisms of heme oxygenase-1-mediated cardiac and pulmonary vascular protection in chronic hypoxia: roles of carbon monoxide and bilirubin. Bilirubin 131-140 heme oxygenase 1 Homo sapiens 14-30 16313248-1 2005 BACKGROUND: A functional GT dinucleotide length polymorphism in the haem oxygenase-1 (HO-1) gene promoter is thought to be involved in the pathogenesis of cardiovascular disease. Dinucleoside Phosphates 28-40 heme oxygenase 1 Homo sapiens 68-90 16234431-6 2005 When HO-1 was induced by hemin in vitro, a significant dose-dependent inhibition of osteoclastogenesis was observed. Hemin 25-30 heme oxygenase 1 Homo sapiens 5-9 16234431-12 2005 Moreover, serum levels of bilirubin, a metabolite of HO-1, were elevated in rheumatoid arthritis patients without bone damage, suggesting HO-1 affects bone loss in humans. Bilirubin 26-35 heme oxygenase 1 Homo sapiens 53-57 16234431-12 2005 Moreover, serum levels of bilirubin, a metabolite of HO-1, were elevated in rheumatoid arthritis patients without bone damage, suggesting HO-1 affects bone loss in humans. Bilirubin 26-35 heme oxygenase 1 Homo sapiens 138-142 16308001-4 2005 The aim of our study was to induce heme oxygenase 1 by using hemin in human internal thoracic and radial arteries and to evaluate the effect of this induction on the contractility of these arterial grafts. Hemin 61-66 heme oxygenase 1 Homo sapiens 35-51 16308001-11 2005 Immunohistochemical staining revealed a large expression of heme oxygenase 1 in all vascular layers of hemin-treated internal thoracic artery and radial artery rings. Hemin 103-108 heme oxygenase 1 Homo sapiens 60-76 16308001-12 2005 Enzyme-linked immunosorbent assay studies showed a significant increase in heme oxygenase 1 levels in hemin-treated internal thoracic artery and radial artery rings. Hemin 102-107 heme oxygenase 1 Homo sapiens 75-91 16308001-13 2005 CONCLUSION: Hemin caused in vitro induction of heme oxygenase 1 in human internal thoracic artery and radial artery grafts. Hemin 12-17 heme oxygenase 1 Homo sapiens 47-63 16283523-6 2005 Real-time RT-PCR confirmed the effect of copper on the levels of MT2A, HSPA1A, CYP1A1 and HMOX1 expression. Copper 41-47 heme oxygenase 1 Homo sapiens 90-95 16198234-2 2005 Heme oxygenase-1 (HO-1), implicated in a role in NO resistance, might confer its protective effect through the direct products biliverdin and CO or the secondary product bilirubin. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 18-22 16198234-2 2005 Heme oxygenase-1 (HO-1), implicated in a role in NO resistance, might confer its protective effect through the direct products biliverdin and CO or the secondary product bilirubin. Bilirubin 170-179 heme oxygenase 1 Homo sapiens 0-16 16198234-2 2005 Heme oxygenase-1 (HO-1), implicated in a role in NO resistance, might confer its protective effect through the direct products biliverdin and CO or the secondary product bilirubin. Bilirubin 170-179 heme oxygenase 1 Homo sapiens 18-22 16123320-10 2005 Pretreatment with SB203580, a p38 mitogen-activated protein kinase inhibitor, reduced ALA-induced HO-1 mRNA expression by 75% and inhibited ALA-induced Nrf2 binding to the HO-1 ARE. Thioctic Acid 86-89 heme oxygenase 1 Homo sapiens 98-102 16123320-10 2005 Pretreatment with SB203580, a p38 mitogen-activated protein kinase inhibitor, reduced ALA-induced HO-1 mRNA expression by 75% and inhibited ALA-induced Nrf2 binding to the HO-1 ARE. Thioctic Acid 86-89 heme oxygenase 1 Homo sapiens 172-176 16123320-0 2005 Alpha-lipoic acid-induced heme oxygenase-1 expression is mediated by nuclear factor erythroid 2-related factor 2 and p38 mitogen-activated protein kinase in human monocytic cells. Thioctic Acid 0-17 heme oxygenase 1 Homo sapiens 26-42 16123320-10 2005 Pretreatment with SB203580, a p38 mitogen-activated protein kinase inhibitor, reduced ALA-induced HO-1 mRNA expression by 75% and inhibited ALA-induced Nrf2 binding to the HO-1 ARE. Thioctic Acid 140-143 heme oxygenase 1 Homo sapiens 172-176 16123320-5 2005 This study examined the effects of ALA on HO-1 expression in human monocytic cells. Thioctic Acid 35-38 heme oxygenase 1 Homo sapiens 42-46 16123320-6 2005 METHODS AND RESULTS: ALA time and dose-dependently induced HO-1 mRNA expression in THP-1 cells, with peak expression at 4 hours and returning to baseline by 24 hours. Thioctic Acid 21-24 heme oxygenase 1 Homo sapiens 59-63 16123320-11 2005 CONCLUSIONS: These results demonstrate that ALA induces HO-1 expression in THP-1 monocytic cells via Nrf2 and p38. Thioctic Acid 44-47 heme oxygenase 1 Homo sapiens 56-60 16123320-8 2005 ALA stimulated translocation of the transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) into the nucleus and binding to a human HO-1 antioxidant response element (ARE) by 30 minutes. Thioctic Acid 0-3 heme oxygenase 1 Homo sapiens 148-152 16123320-12 2005 Further studies are required to investigate whether the protective effects of ALA in monocytes are mediated by HO-1. Thioctic Acid 78-81 heme oxygenase 1 Homo sapiens 111-115 16123320-9 2005 A dominant-negative Nrf2 inhibitor reduced ALA-induced HO-1 mRNA expression by 66%. Thioctic Acid 43-46 heme oxygenase 1 Homo sapiens 55-59 16177082-8 2005 Ro-31-8220, a pan-protein kinase C (PKC) inhibitor, and Go6976, a classical PKC inhibitor, blunted LPS-induced HO-1 mRNA expression in monocytes and THP-1 cells. Ro 31-8220 0-10 heme oxygenase 1 Homo sapiens 111-115 16123320-10 2005 Pretreatment with SB203580, a p38 mitogen-activated protein kinase inhibitor, reduced ALA-induced HO-1 mRNA expression by 75% and inhibited ALA-induced Nrf2 binding to the HO-1 ARE. SB 203580 18-26 heme oxygenase 1 Homo sapiens 98-102 16123320-10 2005 Pretreatment with SB203580, a p38 mitogen-activated protein kinase inhibitor, reduced ALA-induced HO-1 mRNA expression by 75% and inhibited ALA-induced Nrf2 binding to the HO-1 ARE. SB 203580 18-26 heme oxygenase 1 Homo sapiens 172-176 16183497-2 2005 Heme oxygenase-1 (HO-1) has recently been shown to be cytoprotective, and is known to be induced by heme moieties. Heme 100-104 heme oxygenase 1 Homo sapiens 0-16 16183497-2 2005 Heme oxygenase-1 (HO-1) has recently been shown to be cytoprotective, and is known to be induced by heme moieties. Heme 100-104 heme oxygenase 1 Homo sapiens 18-22 16183497-3 2005 We investigated the effects of this hemoglobin-based oxygen carrier on HO-1 induction and proinflammatory activation of pulmonary endothelium. Oxygen 53-59 heme oxygenase 1 Homo sapiens 71-75 16210136-2 2005 Inducibility of this enzyme is modulated by a (GT)n dinucleotide length polymorphism in the HO-1 gene promoter. Dinucleoside Phosphates 52-64 heme oxygenase 1 Homo sapiens 92-96 16210136-6 2005 The HO-1 genotype was assessed using genomic DNA isolated from paraffin-embedded allograft biopsy specimens. Paraffin 63-71 heme oxygenase 1 Homo sapiens 4-8 16177082-8 2005 Ro-31-8220, a pan-protein kinase C (PKC) inhibitor, and Go6976, a classical PKC inhibitor, blunted LPS-induced HO-1 mRNA expression in monocytes and THP-1 cells. Go 6976 56-62 heme oxygenase 1 Homo sapiens 111-115 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Heme 34-38 heme oxygenase 1 Homo sapiens 0-16 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Heme 34-38 heme oxygenase 1 Homo sapiens 18-22 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Bilirubin 72-81 heme oxygenase 1 Homo sapiens 0-16 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Bilirubin 72-81 heme oxygenase 1 Homo sapiens 18-22 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Iron 83-87 heme oxygenase 1 Homo sapiens 0-16 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Iron 83-87 heme oxygenase 1 Homo sapiens 18-22 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 0-16 16249618-2 2005 Heme oxygenase-1 (HO-1) catalyzes heme breakdown, eventually generating bilirubin, iron and carbon monoxide. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 18-22 16408059-4 2005 We further find that nitrite readily affects cyclic GMP production, cytochrome P450 activities, and heat shock protein 70 and heme oxygenase-1 expression in a variety of tissues. Nitrites 21-28 heme oxygenase 1 Homo sapiens 126-142 16309584-0 2005 Heme oxygenase-1-dependent and -independent regulation of angiogenic genes expression: effect of cobalt protoporphyrin and cobalt chloride on VEGF and IL-8 synthesis in human microvascular endothelial cells. cobaltiprotoporphyrin 97-118 heme oxygenase 1 Homo sapiens 0-16 16309584-0 2005 Heme oxygenase-1-dependent and -independent regulation of angiogenic genes expression: effect of cobalt protoporphyrin and cobalt chloride on VEGF and IL-8 synthesis in human microvascular endothelial cells. cobaltous chloride 123-138 heme oxygenase 1 Homo sapiens 0-16 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Heme 33-37 heme oxygenase 1 Homo sapiens 0-4 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltiprotoporphyrin 84-105 heme oxygenase 1 Homo sapiens 13-29 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 43-58 heme oxygenase 1 Homo sapiens 0-4 16309588-2 2005 We previously revealed a substantial oxidation of plasma hemoglobin to methemoglobin and a subsequent heme-catalyzed LDL oxidation generating moieties toxic to endothelium in heme oxygenase-1 (HO-1)-deficiency in human. Heme 102-106 heme oxygenase 1 Homo sapiens 175-191 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltiprotoporphyrin 84-105 heme oxygenase 1 Homo sapiens 31-35 16309588-6 2005 Both cytotoxicity and HO-1 inducing ability of the oxidized LDL were strongly dependent on its lipid hydroperoxide content. Lipid Peroxides 95-114 heme oxygenase 1 Homo sapiens 22-26 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Carbon Monoxide 60-62 heme oxygenase 1 Homo sapiens 0-4 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Iron 65-69 heme oxygenase 1 Homo sapiens 0-4 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). coppix 107-113 heme oxygenase 1 Homo sapiens 13-29 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Biliverdine 75-85 heme oxygenase 1 Homo sapiens 0-4 16309585-2 2005 HO-1 catalyzes the conversion of heme into carbon monoxide (CO), iron, and biliverdin, which is subsequently converted to bilirubin. Bilirubin 122-131 heme oxygenase 1 Homo sapiens 0-4 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). coppix 107-113 heme oxygenase 1 Homo sapiens 31-35 16309585-3 2005 The beneficial effects of HO-1 induction include decreasing pro-oxidants (heme), increasing anti-oxidants (biliverdin and bilirubin), and producing a vasodilator with anti-apoptotic and anti-inflammatory properties (CO). Biliverdine 107-117 heme oxygenase 1 Homo sapiens 26-30 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltous chloride 118-133 heme oxygenase 1 Homo sapiens 13-29 16309585-3 2005 The beneficial effects of HO-1 induction include decreasing pro-oxidants (heme), increasing anti-oxidants (biliverdin and bilirubin), and producing a vasodilator with anti-apoptotic and anti-inflammatory properties (CO). Bilirubin 122-131 heme oxygenase 1 Homo sapiens 26-30 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltous chloride 118-133 heme oxygenase 1 Homo sapiens 31-35 16309586-5 2005 Overexpression of HO-1 decreased both 8-epi-PGF2alpha and MCP-1. 8-epi-prostaglandin F2alpha 38-53 heme oxygenase 1 Homo sapiens 18-22 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltous chloride 135-140 heme oxygenase 1 Homo sapiens 13-29 16309584-1 2005 Induction of heme oxygenase-1 (HO-1) expression can be achieved by stimulation with cobalt protoporphyrin (CoPPIX) or cobalt chloride (CoCl2). cobaltous chloride 135-140 heme oxygenase 1 Homo sapiens 31-35 16309584-2 2005 HO-1 has been recently implicated in regulation of angiogenesis and CoCl2 is known to potently activate hypoxia inducible factor-1 (HIF-1) transcription factor, a key regulator of angiogenic response in hypoxia. cobaltous chloride 68-73 heme oxygenase 1 Homo sapiens 0-4 16309584-4 2005 CoPPIX induced HO-1 expression and strongly enhanced VEGF and IL-8 synthesis, through the activation of VEGF and IL-8 promoters. coppix 0-6 heme oxygenase 1 Homo sapiens 15-19 16309584-8 2005 Our data show that although both CoCl2 and CoPPIX induce HO-1, the influence of CoCl2 on VEGF does not involve HO-1 and is HIF-1-dependent, while the effect of CoPPIX does not involve HIF-1 but relies on HO-1. cobaltous chloride 33-38 heme oxygenase 1 Homo sapiens 57-61 16309584-8 2005 Our data show that although both CoCl2 and CoPPIX induce HO-1, the influence of CoCl2 on VEGF does not involve HO-1 and is HIF-1-dependent, while the effect of CoPPIX does not involve HIF-1 but relies on HO-1. coppix 43-49 heme oxygenase 1 Homo sapiens 57-61 15901614-4 2005 The protective effects of heme oxygenase-1 and products of its enzymatic activity, including carbon monoxide, biliverdin and bilirubin, and ferritin, have opened the door to potential therapeutic and disease-monitoring possibilities that one day may be applicable to pulmonary medicine. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 26-42 15901614-4 2005 The protective effects of heme oxygenase-1 and products of its enzymatic activity, including carbon monoxide, biliverdin and bilirubin, and ferritin, have opened the door to potential therapeutic and disease-monitoring possibilities that one day may be applicable to pulmonary medicine. Biliverdine 110-120 heme oxygenase 1 Homo sapiens 26-42 15901614-4 2005 The protective effects of heme oxygenase-1 and products of its enzymatic activity, including carbon monoxide, biliverdin and bilirubin, and ferritin, have opened the door to potential therapeutic and disease-monitoring possibilities that one day may be applicable to pulmonary medicine. Bilirubin 125-134 heme oxygenase 1 Homo sapiens 26-42 15901601-0 2005 Novel lipid mediator aspirin-triggered lipoxin A4 induces heme oxygenase-1 in endothelial cells. Aspirin 21-28 heme oxygenase 1 Homo sapiens 58-74 15985429-5 2005 Both IAB and tBHQ induced antioxidant response element (ARE)-directed green fluorescent protein (GFP) expression in ARE/thymidine kinase GFP HepG2 cells, and both initiated nuclear Nrf2 accumulation and induction of heme oxygenase 1 in HEK293 cells. 2-tert-butylhydroquinone 13-17 heme oxygenase 1 Homo sapiens 216-232 16020746-6 2005 The proposed mechanisms by which HO-1 exerts its cytoprotective effects include its abilities to degrade the pro-oxidative heme, to release biliverdin and subsequently convert it bilirubin, both of which have antioxidant properties, and to generate carbon monoxide, which has antiproliferative and antiinflammatory as well as vasodilatory properties. Heme 123-127 heme oxygenase 1 Homo sapiens 33-37 16020746-6 2005 The proposed mechanisms by which HO-1 exerts its cytoprotective effects include its abilities to degrade the pro-oxidative heme, to release biliverdin and subsequently convert it bilirubin, both of which have antioxidant properties, and to generate carbon monoxide, which has antiproliferative and antiinflammatory as well as vasodilatory properties. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 33-37 16020746-6 2005 The proposed mechanisms by which HO-1 exerts its cytoprotective effects include its abilities to degrade the pro-oxidative heme, to release biliverdin and subsequently convert it bilirubin, both of which have antioxidant properties, and to generate carbon monoxide, which has antiproliferative and antiinflammatory as well as vasodilatory properties. Bilirubin 179-188 heme oxygenase 1 Homo sapiens 33-37 16020746-6 2005 The proposed mechanisms by which HO-1 exerts its cytoprotective effects include its abilities to degrade the pro-oxidative heme, to release biliverdin and subsequently convert it bilirubin, both of which have antioxidant properties, and to generate carbon monoxide, which has antiproliferative and antiinflammatory as well as vasodilatory properties. Carbon Monoxide 249-264 heme oxygenase 1 Homo sapiens 33-37 15920011-1 2005 Heme oxygenase-1 (HO-1) is an intracellular enzyme that degrades heme and inhibits immune responses and inflammation in vivo. Heme 65-69 heme oxygenase 1 Homo sapiens 0-16 15920011-1 2005 Heme oxygenase-1 (HO-1) is an intracellular enzyme that degrades heme and inhibits immune responses and inflammation in vivo. Heme 65-69 heme oxygenase 1 Homo sapiens 18-22 15920011-6 2005 Induction of HO-1 expression with cobalt protoporphyrin (CoPP) in human and rat DCs inhibits lipopolysaccharide (LPS)-induced phenotypic maturation and secretion of proinflammatory cytokines, resulting in the inhibition of alloreactive T-cell proliferation. cobaltiprotoporphyrin 34-55 heme oxygenase 1 Homo sapiens 13-17 15920011-6 2005 Induction of HO-1 expression with cobalt protoporphyrin (CoPP) in human and rat DCs inhibits lipopolysaccharide (LPS)-induced phenotypic maturation and secretion of proinflammatory cytokines, resulting in the inhibition of alloreactive T-cell proliferation. cobaltiprotoporphyrin 57-61 heme oxygenase 1 Homo sapiens 13-17 15920011-8 2005 Reactive oxygen species induced by LPS in DCs were inhibited by induction of HO-1. Reactive Oxygen Species 0-23 heme oxygenase 1 Homo sapiens 77-81 16020495-2 2005 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous antioxidant bilirubin and carbon monoxide, both of which exert antiinflammatory and antiproliferative effects. Bilirubin 84-93 heme oxygenase 1 Homo sapiens 0-16 16020495-2 2005 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous antioxidant bilirubin and carbon monoxide, both of which exert antiinflammatory and antiproliferative effects. Bilirubin 84-93 heme oxygenase 1 Homo sapiens 18-22 16020495-2 2005 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous antioxidant bilirubin and carbon monoxide, both of which exert antiinflammatory and antiproliferative effects. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 0-16 16020495-2 2005 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous antioxidant bilirubin and carbon monoxide, both of which exert antiinflammatory and antiproliferative effects. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 18-22 15901601-0 2005 Novel lipid mediator aspirin-triggered lipoxin A4 induces heme oxygenase-1 in endothelial cells. lipoxin A4 39-49 heme oxygenase 1 Homo sapiens 58-74 15901601-2 2005 Recently, it was reported that aspirin induces heme oxygenase-1 (HO-1) expression on endothelial cells (EC) in a COX-independent manner, what confers protection against prooxidant insults. Aspirin 31-38 heme oxygenase 1 Homo sapiens 47-63 15901601-2 2005 Recently, it was reported that aspirin induces heme oxygenase-1 (HO-1) expression on endothelial cells (EC) in a COX-independent manner, what confers protection against prooxidant insults. Aspirin 31-38 heme oxygenase 1 Homo sapiens 65-69 15901601-4 2005 In this study, we investigated whether an aspirin-triggered lipoxin A(4) stable analog, 15-epi-16-(para-fluoro)-phenoxy-lipoxin A(4) (ATL-1) was able to induce endothelial HO-1. Aspirin 42-49 heme oxygenase 1 Homo sapiens 172-176 15901601-4 2005 In this study, we investigated whether an aspirin-triggered lipoxin A(4) stable analog, 15-epi-16-(para-fluoro)-phenoxy-lipoxin A(4) (ATL-1) was able to induce endothelial HO-1. 15-epi-16-(para-fluoro)-phenoxy-lipoxin a 88-129 heme oxygenase 1 Homo sapiens 172-176 15901601-8 2005 This inhibitory effect of the analog is modulated by HO-1 because it was blocked by SnPPIX, a competitive inhibitor that blocks HO-1 activity. tin protoporphyrin IX 84-90 heme oxygenase 1 Homo sapiens 53-57 15901601-8 2005 This inhibitory effect of the analog is modulated by HO-1 because it was blocked by SnPPIX, a competitive inhibitor that blocks HO-1 activity. tin protoporphyrin IX 84-90 heme oxygenase 1 Homo sapiens 128-132 15996234-7 2005 Postoperatively, serum transaminases were significantly lower and the bile salt secretion was higher in the initial low HO-1 group, compared to the high expression group. Bile Acids and Salts 70-79 heme oxygenase 1 Homo sapiens 120-124 16146339-0 2005 L-methionine reduces oxidant stress in endothelial cells: role of heme oxygenase-1, ferritin, and nitric oxide. Methionine 0-12 heme oxygenase 1 Homo sapiens 66-82 16146339-2 2005 In the present study, L-methionine led to a concentration-dependent induction of the antioxidant proteins heme oxygenase-1 (HO-1) and ferritin in cultured endothelial cells (ECV 304). Methionine 22-34 heme oxygenase 1 Homo sapiens 106-122 16146339-2 2005 In the present study, L-methionine led to a concentration-dependent induction of the antioxidant proteins heme oxygenase-1 (HO-1) and ferritin in cultured endothelial cells (ECV 304). Methionine 22-34 heme oxygenase 1 Homo sapiens 124-128 16146339-5 2005 The antioxidant effect of L-methionine was mimicked by the HO-1 product bilirubin, which suppressed free radical formation almost completely. Methionine 26-38 heme oxygenase 1 Homo sapiens 59-63 16146339-5 2005 The antioxidant effect of L-methionine was mimicked by the HO-1 product bilirubin, which suppressed free radical formation almost completely. Bilirubin 72-81 heme oxygenase 1 Homo sapiens 59-63 16146339-7 2005 These findings demonstrate that L-methionine reduces free radical formation in endothelial cells, possibly through induction of heme oxygenase-1 and ferritin. Methionine 32-44 heme oxygenase 1 Homo sapiens 128-144 15993853-0 2005 Sequential induction of heme oxygenase-1 and manganese superoxide dismutase protects cultured astrocytes against nitric oxide. Nitric Oxide 113-125 heme oxygenase 1 Homo sapiens 24-40 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Heme 84-88 heme oxygenase 1 Homo sapiens 12-28 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Heme 84-88 heme oxygenase 1 Homo sapiens 30-34 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Bilirubin 92-101 heme oxygenase 1 Homo sapiens 12-28 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Bilirubin 92-101 heme oxygenase 1 Homo sapiens 30-34 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Carbon Monoxide 116-131 heme oxygenase 1 Homo sapiens 12-28 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Carbon Monoxide 116-131 heme oxygenase 1 Homo sapiens 30-34 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Carbon Monoxide 133-135 heme oxygenase 1 Homo sapiens 12-28 16117883-1 2005 BACKGROUND: Heme-oxygenase 1 (HO-1) is a rate-limiting enzyme in the degradation of heme to bilirubin, ferritin and carbon monoxide (CO) and may have significant anti-inflammatory function. Carbon Monoxide 133-135 heme oxygenase 1 Homo sapiens 30-34 16455537-3 2005 A length polymorphic variant of the HO-1 gene promoter region, comprising (GT)n dinucleotide repeats, is associated with altered levels of gene transcription: long (= 36 GT) repeats are associated with decreased HO-1. Dinucleoside Phosphates 80-92 heme oxygenase 1 Homo sapiens 36-40 16455537-3 2005 A length polymorphic variant of the HO-1 gene promoter region, comprising (GT)n dinucleotide repeats, is associated with altered levels of gene transcription: long (= 36 GT) repeats are associated with decreased HO-1. Dinucleoside Phosphates 80-92 heme oxygenase 1 Homo sapiens 212-216 15880142-7 2005 CORM-2 or CORM-3 did not cause any evident cytotoxicity and produced an increase in HO-1 expression and heme oxygenase activity; this effect was completely prevented by the thiol donor N-acetylcysteine. Sulfhydryl Compounds 173-178 heme oxygenase 1 Homo sapiens 84-88 15964514-2 2005 In the present study we investigated induction of HO-1 and adaptive increases in reduced glutathione (GSH) in human aortic smooth muscle cells (SMC) in response to moderately oxidized LDL (moxLDL, 100 microg protein/ml, 24 h), a species containing high levels of lipid hydroperoxides. moxldl 189-195 heme oxygenase 1 Homo sapiens 50-54 15964514-3 2005 Expression and activity of HO-1 and GSH levels were elevated to a greater extent by moxLDL than highly oxidized LDL but unaffected by native or acetylated LDL. moxldl 84-90 heme oxygenase 1 Homo sapiens 27-31 15964514-6 2005 Pretreatment of SMC with the antioxidant vitamin C (100 microM, 24 h) attenuated the induction of HO-1 by moxLDL. Ascorbic Acid 41-50 heme oxygenase 1 Homo sapiens 98-102 15964514-6 2005 Pretreatment of SMC with the antioxidant vitamin C (100 microM, 24 h) attenuated the induction of HO-1 by moxLDL. moxldl 106-112 heme oxygenase 1 Homo sapiens 98-102 15964514-8 2005 These findings demonstrate for the first time that activation of PKC, p38(MAPK), JNK, ERK1/2, and Nrf2 by oxidized LDL in human SMC leads to HO-1 induction, constituting an adaptive response against oxidative injury that can be ameliorated by vitamin C. Ascorbic Acid 243-252 heme oxygenase 1 Homo sapiens 141-145 15993334-9 2005 Whereas inhibiting the ERK pathway with the MEK inhibitor PD98059 significantly decreased HNE-mediated ERK phosphorylation, c-Fos protein induction, AP-1 binding, and HO-1 protein induction, inhibition of the ERK pathway had no effect on HNE-induced HO-1 mRNA. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 58-65 heme oxygenase 1 Homo sapiens 167-171 15993334-9 2005 Whereas inhibiting the ERK pathway with the MEK inhibitor PD98059 significantly decreased HNE-mediated ERK phosphorylation, c-Fos protein induction, AP-1 binding, and HO-1 protein induction, inhibition of the ERK pathway had no effect on HNE-induced HO-1 mRNA. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 58-65 heme oxygenase 1 Homo sapiens 250-254 15880142-7 2005 CORM-2 or CORM-3 did not cause any evident cytotoxicity and produced an increase in HO-1 expression and heme oxygenase activity; this effect was completely prevented by the thiol donor N-acetylcysteine. Acetylcysteine 185-201 heme oxygenase 1 Homo sapiens 84-88 15925276-3 2005 The upregulation of HO-1 can be achieved through the use of pharmaceutical agents, such as metalloporphyrins and some HMG-CoA reductase inhibitors. Metalloporphyrins 91-108 heme oxygenase 1 Homo sapiens 20-24 16181113-3 2005 In a prior study, we reported that increasing HO-1 expression by adenoviral gene transfer prior to hemin exposure attenuated oxidative stress and cell death in astrocytes. Hemin 99-104 heme oxygenase 1 Homo sapiens 46-50 16181113-7 2005 Treatment of cortical astrocyte cultures with 1 microM MG-132 resulted in a rapid increase in Nrf2, to a level that was five-fold that of vehicle-treated cultures by 2 h. This was followed by a three to six-fold increase in HO-1 expression that persisted through the 16 h observation period. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 55-61 heme oxygenase 1 Homo sapiens 224-228 16181113-12 2005 These results suggest that increasing HO-1 expression with a proteasome inhibitor protects astrocytes from heme-mediated oxidative injury. Heme 107-111 heme oxygenase 1 Homo sapiens 38-42 15925276-4 2005 Among other agents, atrial natriretic peptide and donors of nitric oxide (NO) are important modulators of the heme-HO system, either through induction of HO-1 or the biological activity of its products. Nitric Oxide 60-72 heme oxygenase 1 Homo sapiens 154-158 15925276-8 2005 In other cardiovascular situations, delivery of human HO-1 to hyperglycemic rats significantly lowers superoxide (O(2)(-)) levels and prevents EC damage and sloughing of vascular EC into the circulation. Superoxides 102-112 heme oxygenase 1 Homo sapiens 54-58 15925276-8 2005 In other cardiovascular situations, delivery of human HO-1 to hyperglycemic rats significantly lowers superoxide (O(2)(-)) levels and prevents EC damage and sloughing of vascular EC into the circulation. Superoxides 114-118 heme oxygenase 1 Homo sapiens 54-58 15920743-5 2005 The results showed that the treatment of differentiated NB cells with PGA1 for a period of 48 hr increased the expression of HO-1 and catalase, decreased the expression of GPx1 and Mn-SOD-2, and did not change the expression of Cu/Zn-SOD-1 as measured by gene array and confirmed by real-time PCR. prostaglandin A1 70-74 heme oxygenase 1 Homo sapiens 125-142 16240585-1 2005 Heme oxygenase 1 (HO-1) catalyses the oxidation of heme to biliverdin, and its expression is induced by oxidative stress. Heme 51-55 heme oxygenase 1 Homo sapiens 0-16 16240585-1 2005 Heme oxygenase 1 (HO-1) catalyses the oxidation of heme to biliverdin, and its expression is induced by oxidative stress. Heme 51-55 heme oxygenase 1 Homo sapiens 18-22 16240585-1 2005 Heme oxygenase 1 (HO-1) catalyses the oxidation of heme to biliverdin, and its expression is induced by oxidative stress. Biliverdine 59-69 heme oxygenase 1 Homo sapiens 0-16 16240585-1 2005 Heme oxygenase 1 (HO-1) catalyses the oxidation of heme to biliverdin, and its expression is induced by oxidative stress. Biliverdine 59-69 heme oxygenase 1 Homo sapiens 18-22 16240585-9 2005 The present study demonstrates a correlation between PAH exposure, as assessed by urinary 1-OHP, and the induction of HO-1 expression. p-Aminohippuric Acid 53-56 heme oxygenase 1 Homo sapiens 118-122 16240585-9 2005 The present study demonstrates a correlation between PAH exposure, as assessed by urinary 1-OHP, and the induction of HO-1 expression. Oxaliplatin 90-95 heme oxygenase 1 Homo sapiens 118-122 15833736-0 2005 Heme oxygenase-1 gene activation by the NAD(P)H oxidase inhibitor 4-(2-aminoethyl) benzenesulfonyl fluoride via a protein kinase B, p38-dependent signaling pathway in monocytes. 4-(2-aminoethyl)benzenesulfonylfluoride 66-107 heme oxygenase 1 Homo sapiens 0-16 15876423-11 2005 These data suggest that induction of HO-1 protein may participate in the protective mechanism of QE on oxidative stress (H(2)O(2))-induced apoptosis, and reduction of intracellular ROS production and mitochondria dysfunction with blocking apoptotic events were involved. ros 181-184 heme oxygenase 1 Homo sapiens 37-41 15876423-12 2005 Differential anti-apoptotic effect between QE and its glycosides RUT and QI via distinct HO-1 protein induction was also delineated. Glycosides 54-64 heme oxygenase 1 Homo sapiens 89-93 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 203-218 heme oxygenase 1 Homo sapiens 24-29 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 203-218 heme oxygenase 1 Homo sapiens 63-67 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 203-218 heme oxygenase 1 Homo sapiens 149-153 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 246-255 heme oxygenase 1 Homo sapiens 24-29 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 246-255 heme oxygenase 1 Homo sapiens 63-67 15896810-13 2005 Among the various HSPs, HSP32, also known as heme oxygenase I (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 246-255 heme oxygenase 1 Homo sapiens 149-153 15876423-0 2005 Quercetin, but not rutin and quercitrin, prevention of H2O2-induced apoptosis via anti-oxidant activity and heme oxygenase 1 gene expression in macrophages. Quercetin 0-9 heme oxygenase 1 Homo sapiens 108-124 15876423-2 2005 Results of Western blotting show that QE but not its glycoside rutin (RUT) and quicitrin-induced HO-1 protein expression in a time- and dose-dependent manner, and HO-1 protein induced by QE was blocked by an addition of cycloheximide or actinomycin D. Cycloheximide 220-233 heme oxygenase 1 Homo sapiens 163-167 15876423-2 2005 Results of Western blotting show that QE but not its glycoside rutin (RUT) and quicitrin-induced HO-1 protein expression in a time- and dose-dependent manner, and HO-1 protein induced by QE was blocked by an addition of cycloheximide or actinomycin D. Dactinomycin 237-250 heme oxygenase 1 Homo sapiens 163-167 15876423-4 2005 Addition of PD98059, but not SB203580 or SP600125, significantly attenuates QE-induced HO-1 protein and mRNA expression associated with blocking the expression of phosphorylated ERKs proteins. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 12-19 heme oxygenase 1 Homo sapiens 87-91 15876423-10 2005 Furthermore, HO-1 catalytic metabolites carbon monoxide (CO), but not Fe(2+), Fe(3+), biliverdin or bilirubin, performed protective effect on cells from H(2)O(2)-induced cell death with an increase in HO-1 protein expression and ERKs protein phosphorylation. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 13-17 15876423-10 2005 Furthermore, HO-1 catalytic metabolites carbon monoxide (CO), but not Fe(2+), Fe(3+), biliverdin or bilirubin, performed protective effect on cells from H(2)O(2)-induced cell death with an increase in HO-1 protein expression and ERKs protein phosphorylation. Carbon Monoxide 40-55 heme oxygenase 1 Homo sapiens 201-205 15876423-10 2005 Furthermore, HO-1 catalytic metabolites carbon monoxide (CO), but not Fe(2+), Fe(3+), biliverdin or bilirubin, performed protective effect on cells from H(2)O(2)-induced cell death with an increase in HO-1 protein expression and ERKs protein phosphorylation. Carbon Monoxide 57-59 heme oxygenase 1 Homo sapiens 13-17 15876423-10 2005 Furthermore, HO-1 catalytic metabolites carbon monoxide (CO), but not Fe(2+), Fe(3+), biliverdin or bilirubin, performed protective effect on cells from H(2)O(2)-induced cell death with an increase in HO-1 protein expression and ERKs protein phosphorylation. Carbon Monoxide 57-59 heme oxygenase 1 Homo sapiens 201-205 15833736-2 2005 Because HO-1 is up-regulated by NAD(P)H oxidase activators such as lipopolysaccharide and 12-O-tetradecanoylphorbol-13-acetate in monocytic cells, we investigated the gene regulation of HO-1 by the chemical NAD(P)H oxidase inhibitor 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF). 4-(2-aminoethyl)benzenesulfonylfluoride 233-274 heme oxygenase 1 Homo sapiens 8-12 15833736-2 2005 Because HO-1 is up-regulated by NAD(P)H oxidase activators such as lipopolysaccharide and 12-O-tetradecanoylphorbol-13-acetate in monocytic cells, we investigated the gene regulation of HO-1 by the chemical NAD(P)H oxidase inhibitor 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF). 4-(2-aminoethyl)benzenesulfonylfluoride 276-281 heme oxygenase 1 Homo sapiens 8-12 15833736-9 2005 Finally, AEBSF- and PKB-dependent induction of HO-1 promoter activity was reduced by simultaneous mutation of an E-box motif (-47/-42) and a cAMP response element/AP-1 element (-664/-657) of the proximal HO-1 gene promoter. Cyclic AMP 141-145 heme oxygenase 1 Homo sapiens 47-51 15833736-1 2005 Heme oxygenase (HO)-1 is the inducible isoform of the rate-limiting enzyme of heme degradation and modulates the inflammatory immune response. Heme 78-82 heme oxygenase 1 Homo sapiens 0-21 15833736-2 2005 Because HO-1 is up-regulated by NAD(P)H oxidase activators such as lipopolysaccharide and 12-O-tetradecanoylphorbol-13-acetate in monocytic cells, we investigated the gene regulation of HO-1 by the chemical NAD(P)H oxidase inhibitor 4-(2-aminoethyl) benzenesulfonyl fluoride (AEBSF). Tetradecanoylphorbol Acetate 90-126 heme oxygenase 1 Homo sapiens 8-12 15930299-0 2005 The synthetic triterpenoids, CDDO and CDDO-imidazolide, are potent inducers of heme oxygenase-1 and Nrf2/ARE signaling. triterpenoids 14-27 heme oxygenase 1 Homo sapiens 79-95 15930299-0 2005 The synthetic triterpenoids, CDDO and CDDO-imidazolide, are potent inducers of heme oxygenase-1 and Nrf2/ARE signaling. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 29-33 heme oxygenase 1 Homo sapiens 79-95 15930299-0 2005 The synthetic triterpenoids, CDDO and CDDO-imidazolide, are potent inducers of heme oxygenase-1 and Nrf2/ARE signaling. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 38-54 heme oxygenase 1 Homo sapiens 79-95 15930299-3 2005 Transfection studies using a series of reporter constructs show that activation of the human HO-1 promoter by the triterpenoids requires an antioxidant response element (ARE), a cyclic AMP response element, and an E Box sequence. triterpenoids 114-127 heme oxygenase 1 Homo sapiens 93-97 15930299-3 2005 Transfection studies using a series of reporter constructs show that activation of the human HO-1 promoter by the triterpenoids requires an antioxidant response element (ARE), a cyclic AMP response element, and an E Box sequence. Cyclic AMP 178-188 heme oxygenase 1 Homo sapiens 93-97 15930299-7 2005 These studies are the first to investigate the induction of the HO-1 and Nrf2/ARE pathways by CDDO and CDDO-Im, and our results suggest that further in vivo studies are needed to explore the chemopreventive and chemotherapeutic potential of the triterpenoids. 2-cyano-3,12-dioxoolean-1,9-dien-28-oic acid 94-98 heme oxygenase 1 Homo sapiens 64-68 15930299-7 2005 These studies are the first to investigate the induction of the HO-1 and Nrf2/ARE pathways by CDDO and CDDO-Im, and our results suggest that further in vivo studies are needed to explore the chemopreventive and chemotherapeutic potential of the triterpenoids. 1-(2-cyano-3,12-dioxooleana-1,9-dien-28-oyl) imidazole 103-110 heme oxygenase 1 Homo sapiens 64-68 15930299-7 2005 These studies are the first to investigate the induction of the HO-1 and Nrf2/ARE pathways by CDDO and CDDO-Im, and our results suggest that further in vivo studies are needed to explore the chemopreventive and chemotherapeutic potential of the triterpenoids. triterpenoids 245-258 heme oxygenase 1 Homo sapiens 64-68 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 47-68 heme oxygenase 1 Homo sapiens 22-26 15681695-5 2005 Increased HO-1 expression in VSMCs leads to increased production of CO and its second messenger cGMP, which are important regulators of vascular tone and paracrine interactions in the vasculature. Carbon Monoxide 68-70 heme oxygenase 1 Homo sapiens 10-14 15681695-5 2005 Increased HO-1 expression in VSMCs leads to increased production of CO and its second messenger cGMP, which are important regulators of vascular tone and paracrine interactions in the vasculature. Cyclic GMP 96-100 heme oxygenase 1 Homo sapiens 10-14 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 47-68 heme oxygenase 1 Homo sapiens 34-38 15932518-5 2005 In vivo, preventive HO-1 induction by CoPP in acute dextran sodium sulphate (DSS)-induced colitis led to a significant down-regulation of colonic inflammation (P < 0.01) with a concomitant reduction in interferon (IFN)-gamma - but unaffected interleukin (IL)-10-secretion by isolated mesenteric lymph nodes (P < 0.01). dss 77-80 heme oxygenase 1 Homo sapiens 20-24 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 47-68 heme oxygenase 1 Homo sapiens 34-38 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 70-74 heme oxygenase 1 Homo sapiens 22-26 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 70-74 heme oxygenase 1 Homo sapiens 34-38 15932518-4 2005 In vitro induction of HO-1 by the HO-1 inducer cobalt protoporphyrin (CoPP) resulted in a dose-dependent down-regulation of caspase-3 activation in HT-29 cells, indicating an anti-apoptotic function of HO-1 in the intestine. cobaltiprotoporphyrin 70-74 heme oxygenase 1 Homo sapiens 34-38 15932518-5 2005 In vivo, preventive HO-1 induction by CoPP in acute dextran sodium sulphate (DSS)-induced colitis led to a significant down-regulation of colonic inflammation (P < 0.01) with a concomitant reduction in interferon (IFN)-gamma - but unaffected interleukin (IL)-10-secretion by isolated mesenteric lymph nodes (P < 0.01). dextran sodium sulphate 52-75 heme oxygenase 1 Homo sapiens 20-24 15911218-0 2005 AZD3582 increases heme oxygenase-1 expression and antioxidant activity in vascular endothelial and gastric mucosal cells. naproxen-n-butyl nitrate 0-7 heme oxygenase 1 Homo sapiens 18-34 15911218-2 2005 Incubation of human endothelial cells (derived from umbilical cord) with AZD3582 (10-100muM) led to increased expression of heme oxygenase (HO)-1 mRNA and protein. naproxen-n-butyl nitrate 73-80 heme oxygenase 1 Homo sapiens 124-145 15911218-5 2005 Pre-treating endothelial cells with AZD3582 at concentrations that were effective at inducing HO-1 also reduced NADPH-dependent production of oxygen radicals. naproxen-n-butyl nitrate 36-43 heme oxygenase 1 Homo sapiens 94-98 15911218-5 2005 Pre-treating endothelial cells with AZD3582 at concentrations that were effective at inducing HO-1 also reduced NADPH-dependent production of oxygen radicals. NADP 112-117 heme oxygenase 1 Homo sapiens 94-98 15911218-5 2005 Pre-treating endothelial cells with AZD3582 at concentrations that were effective at inducing HO-1 also reduced NADPH-dependent production of oxygen radicals. Reactive Oxygen Species 142-157 heme oxygenase 1 Homo sapiens 94-98 15911218-7 2005 When added exogenously to the cells at low micromolar concentrations, the HO-1 metabolite, bilirubin, virtually abolished NADPH-dependent oxidative stress. Bilirubin 91-100 heme oxygenase 1 Homo sapiens 74-78 15911218-7 2005 When added exogenously to the cells at low micromolar concentrations, the HO-1 metabolite, bilirubin, virtually abolished NADPH-dependent oxidative stress. NADP 122-127 heme oxygenase 1 Homo sapiens 74-78 15911218-8 2005 AZD3582-induced blockade of free-radical formation was reversed in the presence of the HO-1 inhibitor, tin protoporphyrin-IX (SnPP). naproxen-n-butyl nitrate 0-7 heme oxygenase 1 Homo sapiens 87-91 15911218-8 2005 AZD3582-induced blockade of free-radical formation was reversed in the presence of the HO-1 inhibitor, tin protoporphyrin-IX (SnPP). tin protoporphyrin IX 103-124 heme oxygenase 1 Homo sapiens 87-91 15911218-8 2005 AZD3582-induced blockade of free-radical formation was reversed in the presence of the HO-1 inhibitor, tin protoporphyrin-IX (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 126-130 heme oxygenase 1 Homo sapiens 87-91 15911218-10 2005 Our results demonstrate that HO-1 is a novel target of AZD3582. naproxen-n-butyl nitrate 55-62 heme oxygenase 1 Homo sapiens 29-33 15927501-4 2005 Heme is catabolised by heme oxygenase 1, anchored in the endoplasmic reticulum membrane. Heme 0-4 heme oxygenase 1 Homo sapiens 23-39 16044631-5 2005 In particular, heme oxygenase-1 (HO-1), the enzyme involved in heme protein metabolism, can provide antioxidant protection through the production of the antioxidant bilirubin. Heme 15-19 heme oxygenase 1 Homo sapiens 33-37 16044631-5 2005 In particular, heme oxygenase-1 (HO-1), the enzyme involved in heme protein metabolism, can provide antioxidant protection through the production of the antioxidant bilirubin. Bilirubin 165-174 heme oxygenase 1 Homo sapiens 15-31 16044631-5 2005 In particular, heme oxygenase-1 (HO-1), the enzyme involved in heme protein metabolism, can provide antioxidant protection through the production of the antioxidant bilirubin. Bilirubin 165-174 heme oxygenase 1 Homo sapiens 33-37 16044631-6 2005 Furthermore, polyamines have been shown to indirectly increase HO-1 content and antioxidant protection. Polyamines 13-23 heme oxygenase 1 Homo sapiens 63-67 16044631-7 2005 The beta2-adrenoceptor agonist clenbuterol has been shown to stimulate polyamine synthesis and by extension, might provide a margin of antioxidant protection through increasing HO-1 content. Clenbuterol 31-42 heme oxygenase 1 Homo sapiens 177-181 15853972-0 2005 The up-regulation of heme oxygenase-1 expression in human gingival fibroblasts stimulated with nicotine. Nicotine 95-103 heme oxygenase 1 Homo sapiens 21-37 15853972-4 2005 OBJECTIVES: The aim of the present study was to investigate the effects of nicotine on the expression of HO-1 protein in cultured human gingival fibroblasts in vitro and further to compare HO-1 expression in gingival tissues obtained from cigarette smokers and non-smokers in vivo. Nicotine 75-83 heme oxygenase 1 Homo sapiens 105-109 15853972-6 2005 In addition, antioxidants catalase, superoxide dismutase (SOD), and N-acetyl-l-cysteine (NAC) were added to test how they modulated the effects on nicotine-induced HO-1 expression. Nicotine 147-155 heme oxygenase 1 Homo sapiens 164-168 15853972-8 2005 RESULTS: The exposure of quiescent human gingival fibroblasts to 10 mm nicotine resulted in the induction of HO-1 protein expression in a time-dependent manner (p < 0.05). Nicotine 71-79 heme oxygenase 1 Homo sapiens 109-113 15853972-9 2005 The addition of glutathione (GSH) precursor NAC inhibited the nicotine-induced HO-1 protein expression (p < 0.05). Glutathione 16-27 heme oxygenase 1 Homo sapiens 79-83 15853972-9 2005 The addition of glutathione (GSH) precursor NAC inhibited the nicotine-induced HO-1 protein expression (p < 0.05). Glutathione 29-32 heme oxygenase 1 Homo sapiens 79-83 15853972-9 2005 The addition of glutathione (GSH) precursor NAC inhibited the nicotine-induced HO-1 protein expression (p < 0.05). Nicotine 62-70 heme oxygenase 1 Homo sapiens 79-83 15853972-13 2005 CONCLUSIONS: Taken together, these results suggest that HO-1 expression is significantly up-regulated in gingival tissues from cigarette smokers, and nicotine may, among other constituents, be responsible for the enhanced HO-1 expression in vivo. Nicotine 150-158 heme oxygenase 1 Homo sapiens 222-226 15853972-14 2005 The regulation of HO-1 expression induced by nicotine is critically dependent on the intracellular GSH concentration. Nicotine 45-53 heme oxygenase 1 Homo sapiens 18-22 15853972-14 2005 The regulation of HO-1 expression induced by nicotine is critically dependent on the intracellular GSH concentration. Glutathione 99-102 heme oxygenase 1 Homo sapiens 18-22 15855051-7 2005 DEM plus LPS caused synergistic induction of heme oxygenase-1 (HO-1), suggesting its role in the inhibitory effects of DEM. diethyl maleate 0-3 heme oxygenase 1 Homo sapiens 45-61 15855051-7 2005 DEM plus LPS caused synergistic induction of heme oxygenase-1 (HO-1), suggesting its role in the inhibitory effects of DEM. diethyl maleate 0-3 heme oxygenase 1 Homo sapiens 63-67 16020198-8 2005 Acute exposure to Cd was found to trigger the upregulation of genes encoding the chaperone proteins HSP90A, HSP27, HSP40, GRP78, HSP72, and HO-1 in S-cells. Cadmium 18-20 heme oxygenase 1 Homo sapiens 140-144 15897578-9 2005 Treatment of the pancreatic cancer cell lines with gemcitabine or radiation strongly induced HO-1 expression. gemcitabine 51-62 heme oxygenase 1 Homo sapiens 93-97 15890016-1 2005 Heme oxygenase isoforms (HO-1/HO-2) catalyze the conversion of heme to carbon monoxide (CO) and bilirubin. Heme 63-67 heme oxygenase 1 Homo sapiens 0-34 15965068-4 2005 To investigate the molecular basis of the renal injury, we evaluated the expression of the stress marker, heme oxygenase-1 (HO-1), in celecoxib-stimulated mesangial cells. Celecoxib 134-143 heme oxygenase 1 Homo sapiens 106-122 15965068-4 2005 To investigate the molecular basis of the renal injury, we evaluated the expression of the stress marker, heme oxygenase-1 (HO-1), in celecoxib-stimulated mesangial cells. Celecoxib 134-143 heme oxygenase 1 Homo sapiens 124-128 15965068-5 2005 We report here that a COX-2 selective NSAID, celecoxib, induced a concentration- and time-dependent increase of HO-1 expression in glomerular mesangial cells. Celecoxib 45-54 heme oxygenase 1 Homo sapiens 112-116 15965068-6 2005 Celecoxib-induced HO-1 protein expression was inhibited by actinomycin D and cycloheximide, suggesting that de novo transcription and translation are required in this process. Celecoxib 0-9 heme oxygenase 1 Homo sapiens 18-22 15965068-6 2005 Celecoxib-induced HO-1 protein expression was inhibited by actinomycin D and cycloheximide, suggesting that de novo transcription and translation are required in this process. Dactinomycin 59-72 heme oxygenase 1 Homo sapiens 18-22 15965068-6 2005 Celecoxib-induced HO-1 protein expression was inhibited by actinomycin D and cycloheximide, suggesting that de novo transcription and translation are required in this process. Cycloheximide 77-90 heme oxygenase 1 Homo sapiens 18-22 15965068-7 2005 N-acetylcysteine, a free radical scavenger, strongly decreased HO-1 expression, suggesting the involvement of reactive oxygen species (ROS). Acetylcysteine 0-16 heme oxygenase 1 Homo sapiens 63-67 15965068-7 2005 N-acetylcysteine, a free radical scavenger, strongly decreased HO-1 expression, suggesting the involvement of reactive oxygen species (ROS). Reactive Oxygen Species 110-133 heme oxygenase 1 Homo sapiens 63-67 15890016-1 2005 Heme oxygenase isoforms (HO-1/HO-2) catalyze the conversion of heme to carbon monoxide (CO) and bilirubin. Carbon Monoxide 71-86 heme oxygenase 1 Homo sapiens 0-34 15965068-7 2005 N-acetylcysteine, a free radical scavenger, strongly decreased HO-1 expression, suggesting the involvement of reactive oxygen species (ROS). Reactive Oxygen Species 135-138 heme oxygenase 1 Homo sapiens 63-67 15890016-1 2005 Heme oxygenase isoforms (HO-1/HO-2) catalyze the conversion of heme to carbon monoxide (CO) and bilirubin. Carbon Monoxide 88-90 heme oxygenase 1 Homo sapiens 0-34 15965068-8 2005 Celecoxib-induced HO-1 expression was attenuated by pretreatment of the cells with SP 600125 (a specific JNK inhibitor), but not SB 203580 (a specific p38 MAPK inhibitor), or PD 98059 (a specific MEK inhibitor). Celecoxib 0-9 heme oxygenase 1 Homo sapiens 18-22 15890016-1 2005 Heme oxygenase isoforms (HO-1/HO-2) catalyze the conversion of heme to carbon monoxide (CO) and bilirubin. Bilirubin 96-105 heme oxygenase 1 Homo sapiens 0-34 15965068-8 2005 Celecoxib-induced HO-1 expression was attenuated by pretreatment of the cells with SP 600125 (a specific JNK inhibitor), but not SB 203580 (a specific p38 MAPK inhibitor), or PD 98059 (a specific MEK inhibitor). pyrazolanthrone 83-92 heme oxygenase 1 Homo sapiens 18-22 15689417-0 2005 Oxidative stress mediates sodium arsenite-induced expression of heme oxygenase-1, monocyte chemoattractant protein-1, and interleukin-6 in vascular smooth muscle cells. sodium arsenite 26-41 heme oxygenase 1 Homo sapiens 64-80 15965068-10 2005 N-acetylcysteine reduced the stimulatory effect of celecoxib on stress kinase activities, suggesting an involvement of JNK in HO-1 expression. Acetylcysteine 0-16 heme oxygenase 1 Homo sapiens 126-130 15965068-10 2005 N-acetylcysteine reduced the stimulatory effect of celecoxib on stress kinase activities, suggesting an involvement of JNK in HO-1 expression. Celecoxib 51-60 heme oxygenase 1 Homo sapiens 126-130 15965068-11 2005 On the other hand, LY 294002, a phosphatidylinositol 3-kinase (PI-3K)-specific inhibitor, prevented the enhancement of HO-1 expression. 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one 19-28 heme oxygenase 1 Homo sapiens 119-123 15965068-13 2005 In conclusion, our data suggest that celecoxib-induced HO-1 expression in glomerular mesangial cells may be mediated by ROS via the JNK-PI-3K cascade. Celecoxib 37-46 heme oxygenase 1 Homo sapiens 55-59 15965068-13 2005 In conclusion, our data suggest that celecoxib-induced HO-1 expression in glomerular mesangial cells may be mediated by ROS via the JNK-PI-3K cascade. Reactive Oxygen Species 120-123 heme oxygenase 1 Homo sapiens 55-59 15855326-1 2005 Treatment of animals or certain cells with carbon monoxide (CO), a product of heme degradation by heme oxygenase-1 (HO-1), has potent anti-inflammatory and antiapoptotic effects that contribute to the survival of transplanted organs. Carbon Monoxide 43-58 heme oxygenase 1 Homo sapiens 98-114 15855326-1 2005 Treatment of animals or certain cells with carbon monoxide (CO), a product of heme degradation by heme oxygenase-1 (HO-1), has potent anti-inflammatory and antiapoptotic effects that contribute to the survival of transplanted organs. Carbon Monoxide 43-58 heme oxygenase 1 Homo sapiens 116-120 15855326-1 2005 Treatment of animals or certain cells with carbon monoxide (CO), a product of heme degradation by heme oxygenase-1 (HO-1), has potent anti-inflammatory and antiapoptotic effects that contribute to the survival of transplanted organs. Carbon Monoxide 60-62 heme oxygenase 1 Homo sapiens 98-114 15855326-1 2005 Treatment of animals or certain cells with carbon monoxide (CO), a product of heme degradation by heme oxygenase-1 (HO-1), has potent anti-inflammatory and antiapoptotic effects that contribute to the survival of transplanted organs. Carbon Monoxide 60-62 heme oxygenase 1 Homo sapiens 116-120 15843512-4 2005 In this study, we present a model for Treg activity that implicates carbon monoxide, a by-product of heme oxygenase-1 activity, as an important and underappreciated facet in the suppressive capacity of Treg. Carbon Monoxide 68-83 heme oxygenase 1 Homo sapiens 101-117 15689417-8 2005 Furthermore, using tin protoporphyrin IX (SnPP) and anti-MCP-1 antibody to abolish iAs-induced HO-1 and MCP-1 activity, respectively, shows that HO-1 has protective effect against iAs-induced injury in VSMCs and MCP-1 is chemoattractive to human monocytes, THP-1. S-Nitroso-N-propionyl-D,L-penicillamine 42-46 heme oxygenase 1 Homo sapiens 95-99 15689417-8 2005 Furthermore, using tin protoporphyrin IX (SnPP) and anti-MCP-1 antibody to abolish iAs-induced HO-1 and MCP-1 activity, respectively, shows that HO-1 has protective effect against iAs-induced injury in VSMCs and MCP-1 is chemoattractive to human monocytes, THP-1. S-Nitroso-N-propionyl-D,L-penicillamine 42-46 heme oxygenase 1 Homo sapiens 145-149 15661856-1 2005 Heme oxygenase (HO)-1 has been shown to be an important biological target of nitric oxide (NO). Nitric Oxide 77-89 heme oxygenase 1 Homo sapiens 0-21 15834314-2 2005 The inducible form of heme oxygenase-1 (HO-1) can be induced by cytokines, lipopolysaccharide, and reactive oxygen species during sepsis. Reactive Oxygen Species 99-122 heme oxygenase 1 Homo sapiens 22-38 15834314-2 2005 The inducible form of heme oxygenase-1 (HO-1) can be induced by cytokines, lipopolysaccharide, and reactive oxygen species during sepsis. Reactive Oxygen Species 99-122 heme oxygenase 1 Homo sapiens 40-44 15689417-6 2005 In iAs-treated rVSMCs, catalase, dimethylsulfoxide, and L-omega-nitro-L-arginine significantly inhibited the increase in expression of all three genes, allopurinol inhibited the increase in MCP-1 and IL-6 expression, but had no effect on HO-1 expression, while superoxide dismutase had no significant effect on HO-1 expression, but had an inhibitory effect on IL-6 expression and a stimulatory effect on MCP-1 expression. l-omega-nitro-l-arginine 56-80 heme oxygenase 1 Homo sapiens 238-242 15834587-0 2005 Heme oxygenase-1 accelerates protumoral effects of nitric oxide in cancer cells. Nitric Oxide 51-63 heme oxygenase 1 Homo sapiens 0-16 15834587-1 2005 We examined the biological effects of nitric oxide (NO) and its mediator, heme oxygenase-1 (HO-1), in cancer. Nitric Oxide 38-50 heme oxygenase 1 Homo sapiens 92-96 15834587-10 2005 HO-1 inhibition by HO-1 antisense S-oligodeoxynucleotide treatment increased NO-induced growth inhibition, and decreased Bcl-2 expression or VEGF secretion in the three cell lines. Oligodeoxyribonucleotides 36-56 heme oxygenase 1 Homo sapiens 0-4 15689417-6 2005 In iAs-treated rVSMCs, catalase, dimethylsulfoxide, and L-omega-nitro-L-arginine significantly inhibited the increase in expression of all three genes, allopurinol inhibited the increase in MCP-1 and IL-6 expression, but had no effect on HO-1 expression, while superoxide dismutase had no significant effect on HO-1 expression, but had an inhibitory effect on IL-6 expression and a stimulatory effect on MCP-1 expression. l-omega-nitro-l-arginine 56-80 heme oxygenase 1 Homo sapiens 311-315 15834587-10 2005 HO-1 inhibition by HO-1 antisense S-oligodeoxynucleotide treatment increased NO-induced growth inhibition, and decreased Bcl-2 expression or VEGF secretion in the three cell lines. Oligodeoxyribonucleotides 36-56 heme oxygenase 1 Homo sapiens 19-23 15689417-7 2005 Therefore, iAs may enhance the expression of HO-1, MCP-1, and IL-6 in VSMCs via different reactive oxygen molecules. 4-Iodoacetamidosalicylic acid 11-14 heme oxygenase 1 Homo sapiens 45-49 15689417-8 2005 Furthermore, using tin protoporphyrin IX (SnPP) and anti-MCP-1 antibody to abolish iAs-induced HO-1 and MCP-1 activity, respectively, shows that HO-1 has protective effect against iAs-induced injury in VSMCs and MCP-1 is chemoattractive to human monocytes, THP-1. tin protoporphyrin IX 19-40 heme oxygenase 1 Homo sapiens 95-99 15689417-8 2005 Furthermore, using tin protoporphyrin IX (SnPP) and anti-MCP-1 antibody to abolish iAs-induced HO-1 and MCP-1 activity, respectively, shows that HO-1 has protective effect against iAs-induced injury in VSMCs and MCP-1 is chemoattractive to human monocytes, THP-1. tin protoporphyrin IX 19-40 heme oxygenase 1 Homo sapiens 145-149 15770161-0 2005 Heme oxygenase-1 induction by dieldrin in dopaminergic cells. Dieldrin 30-38 heme oxygenase 1 Homo sapiens 0-16 15741166-4 2005 Presently, small interference (si) RNA constructs were used to investigate the role of human BVR in sodium arsenite (As)-mediated induction of HO-1 and in cytoprotection against apoptosis. sodium arsenite 100-115 heme oxygenase 1 Homo sapiens 143-147 15741166-4 2005 Presently, small interference (si) RNA constructs were used to investigate the role of human BVR in sodium arsenite (As)-mediated induction of HO-1 and in cytoprotection against apoptosis. arsenite 117-119 heme oxygenase 1 Homo sapiens 143-147 15770161-1 2005 We investigated the transcriptional events and signaling pathways involved in the induction of heme oxygenase-1 (HO-1) by dieldrin, an environmental risk factor of Parkinson"s disease, in a dopaminergic neuronal cells (SN4741). Dieldrin 122-130 heme oxygenase 1 Homo sapiens 95-111 15770161-1 2005 We investigated the transcriptional events and signaling pathways involved in the induction of heme oxygenase-1 (HO-1) by dieldrin, an environmental risk factor of Parkinson"s disease, in a dopaminergic neuronal cells (SN4741). Dieldrin 122-130 heme oxygenase 1 Homo sapiens 113-117 15770161-2 2005 Dieldrin exposure caused dose-dependent and time-dependent induction of heme oxygenase activity and HO-1 protein expression. Dieldrin 0-8 heme oxygenase 1 Homo sapiens 100-104 15770161-3 2005 Deletional and mutational analyses showed that the 5" distal enhancers, E1 and E2, mediate dieldrin-induced HO-1 gene transcription, and the AP-1 DNA binding sites in the E2 enhancer are critical for E2-mediated HO-1 gene activation. Dieldrin 91-99 heme oxygenase 1 Homo sapiens 108-112 15770161-3 2005 Deletional and mutational analyses showed that the 5" distal enhancers, E1 and E2, mediate dieldrin-induced HO-1 gene transcription, and the AP-1 DNA binding sites in the E2 enhancer are critical for E2-mediated HO-1 gene activation. Dieldrin 91-99 heme oxygenase 1 Homo sapiens 212-216 15770161-4 2005 Furthermore, both the p38 and JNK mitogen-activated protein kinase pathways are utilized for HO-1 transcriptional activation by dieldrin. Dieldrin 128-136 heme oxygenase 1 Homo sapiens 93-97 15770161-5 2005 HO-1 inhibitor, ZnPP IX reduced the expression of HO-1 but enhanced the cytotoxicity induced by dieldrin. zinc protoporphyrin 16-20 heme oxygenase 1 Homo sapiens 50-54 15770161-5 2005 HO-1 inhibitor, ZnPP IX reduced the expression of HO-1 but enhanced the cytotoxicity induced by dieldrin. Dieldrin 96-104 heme oxygenase 1 Homo sapiens 0-4 15763346-8 2005 Moreover, NASH patients with lower levels of GSH exhibited higher expression of HO-1. Glutathione 45-48 heme oxygenase 1 Homo sapiens 80-84 15869055-2 2005 These products have important physiologic effects: bilirubin is a potent antioxidant that can act against ischemia/reperfusion injury; there is a negative correlation between the content of HO-1 and the incidence of coronary heart disease (CHD). Bilirubin 51-60 heme oxygenase 1 Homo sapiens 190-194 16181106-0 2005 Propofol inhibits caspase-3 in astroglial cells: role of heme oxygenase-1. Propofol 0-8 heme oxygenase 1 Homo sapiens 57-73 16181102-1 2005 Heme oxygenase (HO-1) is a stress protein, which has been suggested to participate in defense mechanisms against glucose induced oxidative injury. Glucose 113-120 heme oxygenase 1 Homo sapiens 16-20 16181102-2 2005 The purpose of this study was to examine the role of human HO-1 in attenuating glucose-mediated oxidative stress. Glucose 79-86 heme oxygenase 1 Homo sapiens 59-63 16181102-3 2005 We investigated the effect of high ambient glucose (15, 33 and 66 mM) on HO-1 gene expression in endothelial cells grown in a serum deprived media compared to the effect of glucose on exponentially grown cells (10% FBS). Glucose 43-50 heme oxygenase 1 Homo sapiens 73-77 16181102-4 2005 High glucose at 15 and 33 mM caused significant inhibition of HO-1 protein and activity in G0/G1 and in cells exponentially grown. Glucose 5-12 heme oxygenase 1 Homo sapiens 62-66 16181102-5 2005 Glucose concentration at 66 mM caused a significant increase in HO-1. Glucose 0-7 heme oxygenase 1 Homo sapiens 64-68 16181102-6 2005 Addition of heme (10 microM) increased HO-1 protein and bilirubin formation in G0/G1, in a time dependent manner peaking at 16 h. Glucose attenuated heme mediated increase in HO-1 proteins. Heme 12-16 heme oxygenase 1 Homo sapiens 39-43 16181102-6 2005 Addition of heme (10 microM) increased HO-1 protein and bilirubin formation in G0/G1, in a time dependent manner peaking at 16 h. Glucose attenuated heme mediated increase in HO-1 proteins. Heme 12-16 heme oxygenase 1 Homo sapiens 175-179 16181102-7 2005 RT-PCR demonstrated that glucose decreased the levels of HO-1 mRNA in both G0/G1 or cells grown in 10% FBS. Glucose 25-32 heme oxygenase 1 Homo sapiens 57-61 16181102-8 2005 The rate of HO-1 induction in response to heme was several fold higher in serum-starved cells compared to cells cultured in 10% FBS. Heme 42-46 heme oxygenase 1 Homo sapiens 12-16 16181102-10 2005 HO-1 gene transduction prevented glucose-mediated elevation of 8-epi-isoprostane PGF(2alpha). Glucose 33-40 heme oxygenase 1 Homo sapiens 0-4 16181102-10 2005 HO-1 gene transduction prevented glucose-mediated elevation of 8-epi-isoprostane PGF(2alpha). 8-epi-isoprostane 63-80 heme oxygenase 1 Homo sapiens 0-4 16181102-10 2005 HO-1 gene transduction prevented glucose-mediated elevation of 8-epi-isoprostane PGF(2alpha). Prostaglandins F 81-84 heme oxygenase 1 Homo sapiens 0-4 16181102-11 2005 These results imply that expression of HO-1 in G0/G1 cells may be a key player in decreasing cellular heme, associated with increased generation of bilirubin, and in attenuating glucose mediated oxidative stress. Heme 102-106 heme oxygenase 1 Homo sapiens 39-43 16181102-11 2005 These results imply that expression of HO-1 in G0/G1 cells may be a key player in decreasing cellular heme, associated with increased generation of bilirubin, and in attenuating glucose mediated oxidative stress. Bilirubin 148-157 heme oxygenase 1 Homo sapiens 39-43 16181102-11 2005 These results imply that expression of HO-1 in G0/G1 cells may be a key player in decreasing cellular heme, associated with increased generation of bilirubin, and in attenuating glucose mediated oxidative stress. Glucose 178-185 heme oxygenase 1 Homo sapiens 39-43 15688187-1 2005 Heme oxygenase-1 (HO-1) acts in cytoprotection against oxidants and aromatic hydrocarbons in cigarette smoke. Hydrocarbons, Aromatic 68-89 heme oxygenase 1 Homo sapiens 0-16 15688187-1 2005 Heme oxygenase-1 (HO-1) acts in cytoprotection against oxidants and aromatic hydrocarbons in cigarette smoke. Hydrocarbons, Aromatic 68-89 heme oxygenase 1 Homo sapiens 18-22 15618017-2 2005 Overexpression of HSF-1 and heat-shock experiments indicated that HSF-1 repressed the 15d-PGJ2-and arsenite-induced HO-1 gene expression through directly binding to the consensus heat shock element (HSE) of the HO-1 gene promoter. 15-deoxy-delta(12,14)-prostaglandin J2 86-94 heme oxygenase 1 Homo sapiens 116-120 15826493-3 2005 SUL strongly induced Nrf2 protein expression and ARE-mediated transcription activation, retarded degradation of Nrf2 through inhibiting Keap1, and thereby activating the transcriptional expression of HO-1. sulforaphane 0-3 heme oxygenase 1 Homo sapiens 200-204 15786820-4 2005 METHODS: Real-time RT-PCR was performed for complement component C3 and heme oxygenase-1 (HO-1) genes compared to the housekeeping gene beta-actin using whole section RNA extracted from formalin-fixed and paraffin-embedded archival material of 16 explanted, rejected renal allografts. Paraffin 205-213 heme oxygenase 1 Homo sapiens 90-94 15618017-2 2005 Overexpression of HSF-1 and heat-shock experiments indicated that HSF-1 repressed the 15d-PGJ2-and arsenite-induced HO-1 gene expression through directly binding to the consensus heat shock element (HSE) of the HO-1 gene promoter. 15-deoxy-delta(12,14)-prostaglandin J2 86-94 heme oxygenase 1 Homo sapiens 211-215 15618017-2 2005 Overexpression of HSF-1 and heat-shock experiments indicated that HSF-1 repressed the 15d-PGJ2-and arsenite-induced HO-1 gene expression through directly binding to the consensus heat shock element (HSE) of the HO-1 gene promoter. arsenite 99-107 heme oxygenase 1 Homo sapiens 116-120 15618017-2 2005 Overexpression of HSF-1 and heat-shock experiments indicated that HSF-1 repressed the 15d-PGJ2-and arsenite-induced HO-1 gene expression through directly binding to the consensus heat shock element (HSE) of the HO-1 gene promoter. arsenite 99-107 heme oxygenase 1 Homo sapiens 211-215 15740980-8 2005 When the GSH-depleted or BSO-pretreated macrophages were exposed to NO, delivered either exogenously from spermine NONOate or endogenously from LPS-derived elevation of iNOS, super-induction of HO-1 was observed. Glutathione 9-12 heme oxygenase 1 Homo sapiens 194-198 15690204-2 2005 Conserved glycines, Gly139 and Gly143, in the distal helix of human heme oxygenase-1 (HO-1) provide the flexibility required for the opening and closing of the heme active site for substrate binding and product dissociation during HO-1 catalysis. Glycine 10-18 heme oxygenase 1 Homo sapiens 68-84 15690204-2 2005 Conserved glycines, Gly139 and Gly143, in the distal helix of human heme oxygenase-1 (HO-1) provide the flexibility required for the opening and closing of the heme active site for substrate binding and product dissociation during HO-1 catalysis. Glycine 10-18 heme oxygenase 1 Homo sapiens 86-90 15690204-2 2005 Conserved glycines, Gly139 and Gly143, in the distal helix of human heme oxygenase-1 (HO-1) provide the flexibility required for the opening and closing of the heme active site for substrate binding and product dissociation during HO-1 catalysis. Glycine 10-18 heme oxygenase 1 Homo sapiens 231-235 15690204-2 2005 Conserved glycines, Gly139 and Gly143, in the distal helix of human heme oxygenase-1 (HO-1) provide the flexibility required for the opening and closing of the heme active site for substrate binding and product dissociation during HO-1 catalysis. Heme 68-72 heme oxygenase 1 Homo sapiens 86-90 15690204-2 2005 Conserved glycines, Gly139 and Gly143, in the distal helix of human heme oxygenase-1 (HO-1) provide the flexibility required for the opening and closing of the heme active site for substrate binding and product dissociation during HO-1 catalysis. Heme 68-72 heme oxygenase 1 Homo sapiens 231-235 15740980-0 2005 Super-induction of HO-1 in macrophages stimulated with lipopolysaccharide by prior depletion of glutathione decreases iNOS expression and NO production. Glutathione 96-107 heme oxygenase 1 Homo sapiens 19-23 15740980-2 2005 In response to the depletion of GSH, expression of HO-1 is induced and HO activity is elevated. Glutathione 32-35 heme oxygenase 1 Homo sapiens 51-55 15740980-6 2005 In support of this, HO-1 is induced in macrophages treated only with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis depleting the GSH level. Buthionine Sulfoximine 69-91 heme oxygenase 1 Homo sapiens 20-24 15740980-6 2005 In support of this, HO-1 is induced in macrophages treated only with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis depleting the GSH level. Buthionine Sulfoximine 93-96 heme oxygenase 1 Homo sapiens 20-24 15740980-6 2005 In support of this, HO-1 is induced in macrophages treated only with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis depleting the GSH level. Glutathione 115-118 heme oxygenase 1 Homo sapiens 20-24 15740980-6 2005 In support of this, HO-1 is induced in macrophages treated only with buthionine sulfoximine (BSO), an inhibitor of GSH biosynthesis depleting the GSH level. Glutathione 146-149 heme oxygenase 1 Homo sapiens 20-24 15740980-7 2005 Alternatively, when the macrophages were exposed to spermine NONOate, an exogenous NO-donor, HO-1, was induced also. spermine nitric oxide complex 52-68 heme oxygenase 1 Homo sapiens 93-97 15740980-8 2005 When the GSH-depleted or BSO-pretreated macrophages were exposed to NO, delivered either exogenously from spermine NONOate or endogenously from LPS-derived elevation of iNOS, super-induction of HO-1 was observed. spermine nitric oxide complex 106-122 heme oxygenase 1 Homo sapiens 194-198 15740980-10 2005 Thus, when the depletion of GSH is combined with NO delivery, expression of HO-1 is enhanced to a greater extent than that enhanced either by GSH depletion or by NO delivery. Glutathione 28-31 heme oxygenase 1 Homo sapiens 76-80 15740980-10 2005 Thus, when the depletion of GSH is combined with NO delivery, expression of HO-1 is enhanced to a greater extent than that enhanced either by GSH depletion or by NO delivery. Glutathione 142-145 heme oxygenase 1 Homo sapiens 76-80 15590657-8 2005 Promoter sequence analysis revealed two putative Ets binding sites (EBSs) in this region, and mutation of these sites showed that EBS -93, more than EBS -125, was critical for full HO-1 promoter activity. ethylbenzene 130-133 heme oxygenase 1 Homo sapiens 181-185 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Heme 141-145 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Heme 141-145 heme oxygenase 1 Homo sapiens 18-22 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Iron 177-181 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Iron 177-181 heme oxygenase 1 Homo sapiens 18-22 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Iron 183-185 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Iron 183-185 heme oxygenase 1 Homo sapiens 18-22 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Biliverdine 188-198 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Biliverdine 188-198 heme oxygenase 1 Homo sapiens 18-22 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Carbon Monoxide 212-227 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Carbon Monoxide 212-227 heme oxygenase 1 Homo sapiens 18-22 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Carbon Monoxide 229-231 heme oxygenase 1 Homo sapiens 0-16 15649645-1 2005 Heme oxygenase-1 (HO-1) is a stress-responsive enzyme that acts during inflammatory reactions as the rate-limiting step in the catabolism of heme, yielding equimolar amounts of iron (Fe), biliverdin, and the gas carbon monoxide (CO). Carbon Monoxide 229-231 heme oxygenase 1 Homo sapiens 18-22 15649645-4 2005 We will argue that the protective effects exerted by HO-1 are mediated to a large extent by the end products that it generates via the catabolism of heme. Heme 149-153 heme oxygenase 1 Homo sapiens 53-57 15590657-9 2005 Additional studies showed that EBS -93 binds Ets-2 and that mutation of the DNA binding domain of Ets-2 entirely prevented transactivation of HO-1. ethylbenzene 31-34 heme oxygenase 1 Homo sapiens 142-146 15466933-4 2005 Treatment with As(2)O(3) and trolox increased intracellular oxidative stress, as evidenced by heme oxygenase-1 (HO-1) protein levels, c-Jun terminal kinase (JNK) activation, and protein and lipid oxidation. Arsenic Trioxide 15-24 heme oxygenase 1 Homo sapiens 94-110 15466933-4 2005 Treatment with As(2)O(3) and trolox increased intracellular oxidative stress, as evidenced by heme oxygenase-1 (HO-1) protein levels, c-Jun terminal kinase (JNK) activation, and protein and lipid oxidation. Arsenic Trioxide 15-24 heme oxygenase 1 Homo sapiens 112-116 15466933-4 2005 Treatment with As(2)O(3) and trolox increased intracellular oxidative stress, as evidenced by heme oxygenase-1 (HO-1) protein levels, c-Jun terminal kinase (JNK) activation, and protein and lipid oxidation. 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid 29-35 heme oxygenase 1 Homo sapiens 94-110 15466933-4 2005 Treatment with As(2)O(3) and trolox increased intracellular oxidative stress, as evidenced by heme oxygenase-1 (HO-1) protein levels, c-Jun terminal kinase (JNK) activation, and protein and lipid oxidation. 6-hydroxy-2,5,7,8-tetramethylchroman-2-carboxylic acid 29-35 heme oxygenase 1 Homo sapiens 112-116 15629867-0 2005 9,10-Phenanthraquinone in diesel exhaust particles downregulates Cu,Zn-SOD and HO-1 in human pulmonary epithelial cells: intracellular iron scavenger 1,10-phenanthroline affords protection against apoptosis. 9,10-phenanthrenequinone 0-22 heme oxygenase 1 Homo sapiens 79-83 15629867-7 2005 Furthermore, treatment of A549 cells with 10-20 microM PQ for 12 h specifically down-regulated protein levels of Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and heme oxygenase-1 (HO-1) by more than 50%. 9,10-phenanthrenequinone 55-57 heme oxygenase 1 Homo sapiens 156-172 15629867-7 2005 Furthermore, treatment of A549 cells with 10-20 microM PQ for 12 h specifically down-regulated protein levels of Cu,Zn-superoxide dismutase (Cu,Zn-SOD) and heme oxygenase-1 (HO-1) by more than 50%. 9,10-phenanthrenequinone 55-57 heme oxygenase 1 Homo sapiens 174-178 15680334-0 2005 N-acetylcysteine attenuates early induction of heme oxygenase-1 following traumatic brain injury. Acetylcysteine 0-16 heme oxygenase 1 Homo sapiens 47-63 15680334-3 2005 We have studied the temporal and spatial effects of the antioxidant N-acetylcysteine (NAC) on HO-1 levels following lateral fluid-percussion injury by immunoblotting and immunohistochemistry. Acetylcysteine 68-84 heme oxygenase 1 Homo sapiens 94-98 15680334-3 2005 We have studied the temporal and spatial effects of the antioxidant N-acetylcysteine (NAC) on HO-1 levels following lateral fluid-percussion injury by immunoblotting and immunohistochemistry. Acetylcysteine 86-89 heme oxygenase 1 Homo sapiens 94-98 15680334-7 2005 The administration of NAC 5 min following TBI resulted in a marked reduction in this widespread induction of HO-1, concomitant with a decrease in the volume of injury in all three brain regions. Acetylcysteine 22-25 heme oxygenase 1 Homo sapiens 109-113 15686476-9 2005 Consistent with a cellular stress response to the Abeta(1-42)-induced oxidative stress, FAEE treatment increases the levels of heme oxygenase-1 and heat shock protein 72, which may be regulated by oxidative stresses in a coordinated manner and play a pivotal role in the cytoprotection of neuronal cells against Abeta(1-42)-induced toxicity. faee 88-92 heme oxygenase 1 Homo sapiens 127-143 15611319-0 2005 Heme-induced heme oxygenase-1 (HO-1) in human monocytes inhibits apoptosis despite caspase-3 up-regulation. Heme 0-4 heme oxygenase 1 Homo sapiens 13-29 15611319-0 2005 Heme-induced heme oxygenase-1 (HO-1) in human monocytes inhibits apoptosis despite caspase-3 up-regulation. Heme 0-4 heme oxygenase 1 Homo sapiens 31-35 15611319-2 2005 We studied the effects of heme oxygenase-1 (HO-1) induced by its substrate hemin on apoptosis, caspase-3 expression and the differentiation of freshly isolated human monocytes. Hemin 75-80 heme oxygenase 1 Homo sapiens 26-42 15611319-2 2005 We studied the effects of heme oxygenase-1 (HO-1) induced by its substrate hemin on apoptosis, caspase-3 expression and the differentiation of freshly isolated human monocytes. Hemin 75-80 heme oxygenase 1 Homo sapiens 44-48 15611319-3 2005 Hemin induced HO-1 in a dose- and time-dependent fashion as measured by semi-quantitative RT-PCR and flow cytometry. Hemin 0-5 heme oxygenase 1 Homo sapiens 14-18 15611319-5 2005 The specific HO-1 inhibitor zinc protoporphyrin (ZnPP) reversed the effects of hemin on monocyte apoptosis and diminished cell lifespan. zinc protoporphyrin 28-47 heme oxygenase 1 Homo sapiens 13-17 15611319-5 2005 The specific HO-1 inhibitor zinc protoporphyrin (ZnPP) reversed the effects of hemin on monocyte apoptosis and diminished cell lifespan. zinc protoporphyrin 49-53 heme oxygenase 1 Homo sapiens 13-17 15525643-1 2005 The ability of the human heme oxygenase-1 (hHO-1) R183E mutant to oxidize heme in reactions supported by either NADPH-cytochrome P450 reductase or ascorbic acid has been compared. Heme 25-29 heme oxygenase 1 Homo sapiens 43-48 15708121-1 2005 Heme oxygenases (HO-1 and HO-2) are responsible for the production of carbon monoxide, a vasodilator. Carbon Monoxide 70-85 heme oxygenase 1 Homo sapiens 0-30 15525643-2 2005 The NADPH-dependent reaction, like that of wild-type hHO-1, yields exclusively biliverdin IXalpha. NADP 4-9 heme oxygenase 1 Homo sapiens 53-58 15525643-2 2005 The NADPH-dependent reaction, like that of wild-type hHO-1, yields exclusively biliverdin IXalpha. Biliverdine 79-97 heme oxygenase 1 Homo sapiens 53-58 15525643-7 2005 The crystal structure of the R183E mutant, determined in the ferric and ferrous-NO bound forms, shows that the heme primarily adopts the same orientation as in wild-type hHO-1. ferrous-no 72-82 heme oxygenase 1 Homo sapiens 170-175 15525643-7 2005 The crystal structure of the R183E mutant, determined in the ferric and ferrous-NO bound forms, shows that the heme primarily adopts the same orientation as in wild-type hHO-1. Heme 111-115 heme oxygenase 1 Homo sapiens 170-175 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Heme 86-90 heme oxygenase 1 Homo sapiens 0-16 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Heme 86-90 heme oxygenase 1 Homo sapiens 18-22 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Biliverdine 94-104 heme oxygenase 1 Homo sapiens 0-16 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Biliverdine 94-104 heme oxygenase 1 Homo sapiens 18-22 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Iron 106-110 heme oxygenase 1 Homo sapiens 0-16 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Iron 106-110 heme oxygenase 1 Homo sapiens 18-22 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 116-131 heme oxygenase 1 Homo sapiens 0-16 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 116-131 heme oxygenase 1 Homo sapiens 18-22 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 133-135 heme oxygenase 1 Homo sapiens 0-16 15546873-2 2005 Heme oxygenase-1 (HO-1) is a cytoprotective protein that catalyzes the degradation of heme to biliverdin, iron, and carbon monoxide (CO). Carbon Monoxide 133-135 heme oxygenase 1 Homo sapiens 18-22 15546873-4 2005 The induction of HO-1 by ER stress was blocked by actinomycin D or cycloheximide and was independent of any changes in HO-1 mRNA stability. Dactinomycin 50-63 heme oxygenase 1 Homo sapiens 17-21 15546873-4 2005 The induction of HO-1 by ER stress was blocked by actinomycin D or cycloheximide and was independent of any changes in HO-1 mRNA stability. Cycloheximide 67-80 heme oxygenase 1 Homo sapiens 17-21 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. 2"-phosphate 137-149 heme oxygenase 1 Homo sapiens 25-29 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. NADP 153-158 heme oxygenase 1 Homo sapiens 25-29 15516695-0 2005 Involvement of NADPH in the interaction between heme oxygenase-1 and cytochrome P450 reductase. NADP 15-20 heme oxygenase 1 Homo sapiens 48-94 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. Arginine 176-179 heme oxygenase 1 Homo sapiens 25-29 15516695-1 2005 Heme oxygenase-1 (HO-1) catalyzes the physiological degradation of heme at the expense of molecular oxygen using electrons donated by NADPH-cytochrome P450 reductase (CPR). Heme 67-71 heme oxygenase 1 Homo sapiens 0-16 15516695-1 2005 Heme oxygenase-1 (HO-1) catalyzes the physiological degradation of heme at the expense of molecular oxygen using electrons donated by NADPH-cytochrome P450 reductase (CPR). Heme 67-71 heme oxygenase 1 Homo sapiens 18-22 15516695-1 2005 Heme oxygenase-1 (HO-1) catalyzes the physiological degradation of heme at the expense of molecular oxygen using electrons donated by NADPH-cytochrome P450 reductase (CPR). Oxygen 5-11 heme oxygenase 1 Homo sapiens 18-22 15516695-2 2005 In this study, we investigated the effect of NADP(H) on the interaction of HO-1 with CPR by surface plasmon resonance. NADP 45-52 heme oxygenase 1 Homo sapiens 75-79 15516695-3 2005 We found that HO-1 associated with CPR more tightly in the presence of NADP(+) (K(D) = 0.5 microm) than in its absence (K(D) = 2.4 microm). NADP 71-78 heme oxygenase 1 Homo sapiens 14-18 15516695-4 2005 The HO-1 mutants, K149A, K149A/K153A, and R185A, showed almost no heme degradation activity with NADPH-CPR, whereas they exhibited activity comparable to that of the wild type when sodium ascorbate was used. Heme 66-70 heme oxygenase 1 Homo sapiens 4-8 15516695-4 2005 The HO-1 mutants, K149A, K149A/K153A, and R185A, showed almost no heme degradation activity with NADPH-CPR, whereas they exhibited activity comparable to that of the wild type when sodium ascorbate was used. Ascorbic Acid 181-197 heme oxygenase 1 Homo sapiens 4-8 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. guanidino 58-67 heme oxygenase 1 Homo sapiens 25-29 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. Arginine 77-80 heme oxygenase 1 Homo sapiens 25-29 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. Phosphates 140-149 heme oxygenase 1 Homo sapiens 25-29 15516695-8 2005 Computer modeling of the HO-1/CPR complex showed that the guanidino group of Arg(185) is located within the hydrogen bonding distance of 2"-phosphate of NADPH, suggesting that Arg(185) contributes to the binding to CPR through an electrostatic interaction with the phosphate group. Hydrogen 108-116 heme oxygenase 1 Homo sapiens 25-29 15516695-10 2005 Thus, Lys(149) and Lys(153) appear to interact with CPR in such a way as to orient the redox partners for optimal electron transfer from FMN of CPR to heme of HO-1. Lysine 6-9 heme oxygenase 1 Homo sapiens 159-163 15516695-10 2005 Thus, Lys(149) and Lys(153) appear to interact with CPR in such a way as to orient the redox partners for optimal electron transfer from FMN of CPR to heme of HO-1. Lysine 19-22 heme oxygenase 1 Homo sapiens 159-163 15516695-10 2005 Thus, Lys(149) and Lys(153) appear to interact with CPR in such a way as to orient the redox partners for optimal electron transfer from FMN of CPR to heme of HO-1. Heme 151-155 heme oxygenase 1 Homo sapiens 159-163 15650398-6 2005 Expression of HO-1 responds to chemical and physical agents that directly or indirectly generate ROS. Reactive Oxygen Species 97-100 heme oxygenase 1 Homo sapiens 14-18 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Bilirubin 199-208 heme oxygenase 1 Homo sapiens 18-22 15933765-4 2005 Induction of HO-1 occurs as an adaptive and beneficial response to several injurious stimuli including heme and this inducible nature of HO-1 signifies its importance in several pathophysiological disease states. Heme 103-107 heme oxygenase 1 Homo sapiens 13-17 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Heme 79-83 heme oxygenase 1 Homo sapiens 0-16 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Heme 79-83 heme oxygenase 1 Homo sapiens 18-22 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Iron 126-130 heme oxygenase 1 Homo sapiens 0-16 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Iron 126-130 heme oxygenase 1 Homo sapiens 18-22 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Carbon Monoxide 132-147 heme oxygenase 1 Homo sapiens 0-16 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Carbon Monoxide 132-147 heme oxygenase 1 Homo sapiens 18-22 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Biliverdine 152-162 heme oxygenase 1 Homo sapiens 0-16 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Biliverdine 152-162 heme oxygenase 1 Homo sapiens 18-22 15933765-1 2005 Heme oxygenase-1 (HO-1) is an enzyme which catalyzes the rate-limiting step in heme degradation resulting in the formation of iron, carbon monoxide and biliverdin, which is subsequently converted to bilirubin by biliverdin reductase. Bilirubin 199-208 heme oxygenase 1 Homo sapiens 0-16 15650398-10 2005 HO-1 protein expression can occur in the lung in response to oxidative stress associated with infection, altered oxygen tension, and inflammatory diseases. Oxygen 113-119 heme oxygenase 1 Homo sapiens 0-4 15650398-7 2005 Depletion of cellular reduced glutathione may act as a signal for HO-1 transcriptional activation. Glutathione 30-41 heme oxygenase 1 Homo sapiens 66-70 15650398-8 2005 Furthermore, antioxidants and metal-chelating compounds can modulate HO-1 expression. Metals 30-35 heme oxygenase 1 Homo sapiens 69-73 15899048-1 2005 Heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, is expressed by macrophages and endothelial cells in response to various stresses. Heme 38-42 heme oxygenase 1 Homo sapiens 0-16 15899048-1 2005 Heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, is expressed by macrophages and endothelial cells in response to various stresses. Heme 38-42 heme oxygenase 1 Homo sapiens 18-22 15899048-2 2005 Because ferritin synthesis is stimulated by Fe2+, which is a product of heme degradation, we examined the relation between HO-1 and ferritin levels in the serum of patients with hemophagocytic syndrome (HPS), adult-onset Still"s disease (ASD), and other diseases that may cause hyperferritinemia. ammonium ferrous sulfate 44-48 heme oxygenase 1 Homo sapiens 123-127 15899039-6 2005 Protection from chondrocyte death conferred by 0.1 mM SNP was mediated by heme oxygenase 1 (HO-1), as was revealed by the increased expression of HO-1 in 0.1 mM SNP pretreated chondrocytes and by the reversal of this protective effect by the HO-1 inhibitor, zinc protoporphyrin. zinc protoporphyrin 258-277 heme oxygenase 1 Homo sapiens 74-90 15899039-6 2005 Protection from chondrocyte death conferred by 0.1 mM SNP was mediated by heme oxygenase 1 (HO-1), as was revealed by the increased expression of HO-1 in 0.1 mM SNP pretreated chondrocytes and by the reversal of this protective effect by the HO-1 inhibitor, zinc protoporphyrin. zinc protoporphyrin 258-277 heme oxygenase 1 Homo sapiens 92-96 15899039-6 2005 Protection from chondrocyte death conferred by 0.1 mM SNP was mediated by heme oxygenase 1 (HO-1), as was revealed by the increased expression of HO-1 in 0.1 mM SNP pretreated chondrocytes and by the reversal of this protective effect by the HO-1 inhibitor, zinc protoporphyrin. zinc protoporphyrin 258-277 heme oxygenase 1 Homo sapiens 146-150 15899039-6 2005 Protection from chondrocyte death conferred by 0.1 mM SNP was mediated by heme oxygenase 1 (HO-1), as was revealed by the increased expression of HO-1 in 0.1 mM SNP pretreated chondrocytes and by the reversal of this protective effect by the HO-1 inhibitor, zinc protoporphyrin. zinc protoporphyrin 258-277 heme oxygenase 1 Homo sapiens 146-150 15589375-0 2005 Distinct protective mechanisms of HO-1 and HO-2 against hydroperoxide-induced cytotoxicity. Hydrogen Peroxide 56-69 heme oxygenase 1 Homo sapiens 34-47 15589375-1 2005 Heme oxygenases (HO-1 and HO-2) catalyze the NADPH-cytochrome P(450) reductase (CPR)-dependent degradation of heme into iron, carbon monoxide, and biliverdin, which is reduced into bilirubin. Iron 120-124 heme oxygenase 1 Homo sapiens 0-30 15588712-0 2005 Mechanism of concentration-dependent induction of heme oxygenase-1 by resveratrol in human aortic smooth muscle cells. Resveratrol 70-81 heme oxygenase 1 Homo sapiens 50-66 15588712-1 2005 Resveratrol-mediated heme oxygenase-1 (HO-1) induction has been shown to occur in primary neuronal cultures and is thought to have potential neuroprotective action. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 21-37 15588712-1 2005 Resveratrol-mediated heme oxygenase-1 (HO-1) induction has been shown to occur in primary neuronal cultures and is thought to have potential neuroprotective action. Resveratrol 0-11 heme oxygenase 1 Homo sapiens 39-43 15588712-3 2005 We attempted to examine resveratrol"s HO-1 inducing potency and its induction regulation in human aortic smooth muscle cells (HASMC). Resveratrol 24-35 heme oxygenase 1 Homo sapiens 38-42 15588712-4 2005 We showed that resveratrol-mediated HO-1 induction occurred in concentration- and time-dependent manners, but only at low concentrations (1-10 microM), and that it was modulated at both the transcription and translation levels. Resveratrol 15-26 heme oxygenase 1 Homo sapiens 36-40 15588712-5 2005 Additionally, the results of our study showed that nuclear factor-kappa B (NF-kappaB) inhibitors eliminated resveratrol-mediated HO-1 induction and promoter activity, and that deletion of NF-kappaB binding sites in the HO-1 promoter region strongly reduced promoter activity, suggesting involvement of the NF-kappaB pathway in HO-1 induction by resveratrol. Resveratrol 108-119 heme oxygenase 1 Homo sapiens 129-133 15588712-5 2005 Additionally, the results of our study showed that nuclear factor-kappa B (NF-kappaB) inhibitors eliminated resveratrol-mediated HO-1 induction and promoter activity, and that deletion of NF-kappaB binding sites in the HO-1 promoter region strongly reduced promoter activity, suggesting involvement of the NF-kappaB pathway in HO-1 induction by resveratrol. Resveratrol 108-119 heme oxygenase 1 Homo sapiens 219-223 15588712-5 2005 Additionally, the results of our study showed that nuclear factor-kappa B (NF-kappaB) inhibitors eliminated resveratrol-mediated HO-1 induction and promoter activity, and that deletion of NF-kappaB binding sites in the HO-1 promoter region strongly reduced promoter activity, suggesting involvement of the NF-kappaB pathway in HO-1 induction by resveratrol. Resveratrol 108-119 heme oxygenase 1 Homo sapiens 219-223 15588712-5 2005 Additionally, the results of our study showed that nuclear factor-kappa B (NF-kappaB) inhibitors eliminated resveratrol-mediated HO-1 induction and promoter activity, and that deletion of NF-kappaB binding sites in the HO-1 promoter region strongly reduced promoter activity, suggesting involvement of the NF-kappaB pathway in HO-1 induction by resveratrol. Resveratrol 345-356 heme oxygenase 1 Homo sapiens 219-223 15588712-5 2005 Additionally, the results of our study showed that nuclear factor-kappa B (NF-kappaB) inhibitors eliminated resveratrol-mediated HO-1 induction and promoter activity, and that deletion of NF-kappaB binding sites in the HO-1 promoter region strongly reduced promoter activity, suggesting involvement of the NF-kappaB pathway in HO-1 induction by resveratrol. Resveratrol 345-356 heme oxygenase 1 Homo sapiens 219-223 15588712-7 2005 Likewise, we showed that resveratrol at concentrations of > or =20 microM blocked the activity of NF-kappaB through suppression of I kappa-B alpha (IkappaBalpha) phosphorylation, which caused inhibition of HO-1 induction. Resveratrol 25-36 heme oxygenase 1 Homo sapiens 209-213 15588712-8 2005 Conversely, resveratrol in a range of 1-10 microM enhanced the phosphorylation and degradation of IkappaBalpha, a key step in NF-kappaB activation, resulting in HO-1 induction. Resveratrol 12-23 heme oxygenase 1 Homo sapiens 161-165 15588712-9 2005 Collectively, we suggest that resveratrol-mediated HO-1 expression occurs, at least in part, through the NF-kappaB pathway, which might contribute to resveratrol"s vascular-protective effect at physiological concentrations after moderate red wine consumption. Resveratrol 30-41 heme oxygenase 1 Homo sapiens 51-55 15588712-9 2005 Collectively, we suggest that resveratrol-mediated HO-1 expression occurs, at least in part, through the NF-kappaB pathway, which might contribute to resveratrol"s vascular-protective effect at physiological concentrations after moderate red wine consumption. Resveratrol 150-161 heme oxygenase 1 Homo sapiens 51-55 15881418-4 2005 We tested the effect of the use of protoporphyrins (CoPP and FePP), powerful inducers of the cytoprotective protein heme-oxygenase 1 (HO-1), on allogeneic islet graft survival. Protoporphyrins 35-50 heme oxygenase 1 Homo sapiens 116-132 15881418-4 2005 We tested the effect of the use of protoporphyrins (CoPP and FePP), powerful inducers of the cytoprotective protein heme-oxygenase 1 (HO-1), on allogeneic islet graft survival. Protoporphyrins 35-50 heme oxygenase 1 Homo sapiens 134-138 15881418-12 2005 Administration of protoporphyrins to the recipients of allogeneic islets also resulted in transient powerful immunosuppression with reduced lymphocyte proliferative responses, increased proportion of regulatory cells (CD4+CD25+), decreased mononuclear cell infiltrating the graft, paralleled by a systemic upregulation of HO-1 expression. Protoporphyrins 18-33 heme oxygenase 1 Homo sapiens 322-326 15589375-1 2005 Heme oxygenases (HO-1 and HO-2) catalyze the NADPH-cytochrome P(450) reductase (CPR)-dependent degradation of heme into iron, carbon monoxide, and biliverdin, which is reduced into bilirubin. Carbon Monoxide 126-141 heme oxygenase 1 Homo sapiens 0-30 15589375-1 2005 Heme oxygenases (HO-1 and HO-2) catalyze the NADPH-cytochrome P(450) reductase (CPR)-dependent degradation of heme into iron, carbon monoxide, and biliverdin, which is reduced into bilirubin. Biliverdine 147-157 heme oxygenase 1 Homo sapiens 0-30 15589375-1 2005 Heme oxygenases (HO-1 and HO-2) catalyze the NADPH-cytochrome P(450) reductase (CPR)-dependent degradation of heme into iron, carbon monoxide, and biliverdin, which is reduced into bilirubin. Bilirubin 181-190 heme oxygenase 1 Homo sapiens 0-30 15589375-8 2005 Results indicate that the HO-1 and HO-2 cells are more resistant than controls to hemin and to the organic tert-butyl hydroperoxide, t-BuOOH. Hemin 82-87 heme oxygenase 1 Homo sapiens 26-39 15589375-8 2005 Results indicate that the HO-1 and HO-2 cells are more resistant than controls to hemin and to the organic tert-butyl hydroperoxide, t-BuOOH. tert-Butylhydroperoxide 107-131 heme oxygenase 1 Homo sapiens 26-39 15589375-8 2005 Results indicate that the HO-1 and HO-2 cells are more resistant than controls to hemin and to the organic tert-butyl hydroperoxide, t-BuOOH. di-tert-butyl peroxide 133-140 heme oxygenase 1 Homo sapiens 26-39 15589375-10 2005 The levels of oxidatively modified proteins of HO-1 and HO-2 cells in response to t-BuOOH toxicity are identical, but the level of oxidatively modified proteins of HO-2 cells is less than that of HO-1 cells in response to H(2)O(2) toxicity. di-tert-butyl peroxide 82-89 heme oxygenase 1 Homo sapiens 47-60 15589375-1 2005 Heme oxygenases (HO-1 and HO-2) catalyze the NADPH-cytochrome P(450) reductase (CPR)-dependent degradation of heme into iron, carbon monoxide, and biliverdin, which is reduced into bilirubin. Heme 110-114 heme oxygenase 1 Homo sapiens 0-30 16196283-6 2005 The relation analysis showed that PBMC HO-1 protein and mRNA levels had significantly negative relation with FEV1%, PEFR, MEFR50%, respectively (r = -0.51-0.89, P < 0.05-0.001, respectively) and a positive relation with COHb and serum total IgE (r = 0.48-0. cohb 223-227 heme oxygenase 1 Homo sapiens 39-43 16259722-13 2005 Induction of HSP32 with diacetylrhein appeared to be more effective and may hold greater promise. diacerein 24-37 heme oxygenase 1 Homo sapiens 13-18 15797262-9 2005 Up-regulated HO-1 expression was accompanied by increase of the numbers of Hoechst-33342 positive MSCs, the reduction of infarct size, and the improvement of cardiac function. bisbenzimide ethoxide trihydrochloride 75-88 heme oxygenase 1 Homo sapiens 13-17 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Heme 71-75 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Oxygen 219-225 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Oxygen 227-229 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Nitrogen 236-244 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Carbon Monoxide 291-306 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Carbon Monoxide 308-310 heme oxygenase 1 Homo sapiens 28-32 16291246-3 2005 The NO-derived induction of HO-1 caused (a) rapid elimination of toxic heme to inhibit lipid peroxidation and to prevent further induction of iNOS, (b) rapid production of bile pigment antioxidants to scavenge reactive oxygen (O2-) and nitrogen (NO) metabolites, and (c) rapid production of carbon monoxide (CO) to inhibit further production of O2- and NO by blocking the activities of NADPH-oxidase and iNOS, respectively. Oxygen 345-347 heme oxygenase 1 Homo sapiens 28-32 16291246-4 2005 Thus, the NO overproduced by the O2- -dependent induction of iNOS expression can scavenge O2- to produce ONOO-, first to kill the invading pathogens and second to enhance the HO-1 expression in macrophages. Oxygen 33-35 heme oxygenase 1 Homo sapiens 175-179 16291246-4 2005 Thus, the NO overproduced by the O2- -dependent induction of iNOS expression can scavenge O2- to produce ONOO-, first to kill the invading pathogens and second to enhance the HO-1 expression in macrophages. Oxygen 90-92 heme oxygenase 1 Homo sapiens 175-179 16291246-4 2005 Thus, the NO overproduced by the O2- -dependent induction of iNOS expression can scavenge O2- to produce ONOO-, first to kill the invading pathogens and second to enhance the HO-1 expression in macrophages. oxido nitrite 105-110 heme oxygenase 1 Homo sapiens 175-179 15773552-1 2005 Previously it was shown that thiol antioxidants are potent inhibitors of the NO-dependent induction of heme oxygenase 1 (HOX-1) gene. Sulfhydryl Compounds 29-34 heme oxygenase 1 Homo sapiens 103-119 15773552-1 2005 Previously it was shown that thiol antioxidants are potent inhibitors of the NO-dependent induction of heme oxygenase 1 (HOX-1) gene. Sulfhydryl Compounds 29-34 heme oxygenase 1 Homo sapiens 121-126 15773552-2 2005 However, the mechanism of HOX-1 gene down-regulation by thiol antioxidants and underlying signaling pathway remain unclear. Sulfhydryl Compounds 56-61 heme oxygenase 1 Homo sapiens 26-31 15773552-3 2005 In this study we have examined, whether the scavenging of reactive oxygen and reactive nitrogen species (ROS and RNS) is the major cause for thiol-mediated suppression of the HOX-1 induction by NO. reactive oxygen and reactive nitrogen species 58-103 heme oxygenase 1 Homo sapiens 175-180 16291250-1 2005 Survival of macrophages, which serve as the first-line defense against invading pathogens by invoking the overproduction of highly toxic peroxynitrite (ONOO-), depends on their ability to maintain the intracellular GSH level and to induce the expression of heme oxygenase-1 (HO-1). Peroxynitrous Acid 137-150 heme oxygenase 1 Homo sapiens 257-273 16291250-1 2005 Survival of macrophages, which serve as the first-line defense against invading pathogens by invoking the overproduction of highly toxic peroxynitrite (ONOO-), depends on their ability to maintain the intracellular GSH level and to induce the expression of heme oxygenase-1 (HO-1). Peroxynitrous Acid 137-150 heme oxygenase 1 Homo sapiens 275-279 16291250-3 2005 However, macrophages can survive the toxicity of ONOO- by replenishing the depleted GSH level and by inducing HO-1 expression. onoo 49-53 heme oxygenase 1 Homo sapiens 110-114 16291250-5 2005 However, in cells surviving the toxicity caused by lower doses of O2-, NO, or ONOO-, the depleted intracellular GSH level was replenished, and HO-1 expression was increased, but not when they were coexposed to an inhibitor of HO-1 activity. Oxygen 66-68 heme oxygenase 1 Homo sapiens 143-147 16291250-5 2005 However, in cells surviving the toxicity caused by lower doses of O2-, NO, or ONOO-, the depleted intracellular GSH level was replenished, and HO-1 expression was increased, but not when they were coexposed to an inhibitor of HO-1 activity. Oxygen 66-68 heme oxygenase 1 Homo sapiens 226-230 16291250-5 2005 However, in cells surviving the toxicity caused by lower doses of O2-, NO, or ONOO-, the depleted intracellular GSH level was replenished, and HO-1 expression was increased, but not when they were coexposed to an inhibitor of HO-1 activity. onoo 78-82 heme oxygenase 1 Homo sapiens 143-147 16291250-5 2005 However, in cells surviving the toxicity caused by lower doses of O2-, NO, or ONOO-, the depleted intracellular GSH level was replenished, and HO-1 expression was increased, but not when they were coexposed to an inhibitor of HO-1 activity. onoo 78-82 heme oxygenase 1 Homo sapiens 226-230 16291250-6 2005 Cells exposed to an inhibitor of GSH synthesis had greater induction of HO-1 expression and survived. Glutathione 33-36 heme oxygenase 1 Homo sapiens 72-76 16291250-7 2005 However, cells exposed to an inhibitor of HO-1 activity died extensively and could not be revived by addition of N-acetylcysteine (NAC), a precursor of GSH synthesis. Glutathione 152-155 heme oxygenase 1 Homo sapiens 42-46 16291250-8 2005 Thus, the dichotomous cytotoxic or cytoprotective effects of O2-, NO, or ONOO- in macrophages are determined both by cellular GSH level and by HO-1 activity. Oxygen 61-63 heme oxygenase 1 Homo sapiens 143-147 16291250-8 2005 Thus, the dichotomous cytotoxic or cytoprotective effects of O2-, NO, or ONOO- in macrophages are determined both by cellular GSH level and by HO-1 activity. onoo 73-77 heme oxygenase 1 Homo sapiens 143-147 15773552-4 2005 Further, to identify the ROS family members implicated in the HOX-1 induction, we also exposed cells to various non-thiol antioxidants: dimethyl sulfoxide, dimetylthiourea, sodium salicylate, sodium formate, uric acid, catalase, and superoxide dismutase. ros 25-28 heme oxygenase 1 Homo sapiens 62-67 15773552-4 2005 Further, to identify the ROS family members implicated in the HOX-1 induction, we also exposed cells to various non-thiol antioxidants: dimethyl sulfoxide, dimetylthiourea, sodium salicylate, sodium formate, uric acid, catalase, and superoxide dismutase. Dimethyl Sulfoxide 136-154 heme oxygenase 1 Homo sapiens 62-67 15773552-4 2005 Further, to identify the ROS family members implicated in the HOX-1 induction, we also exposed cells to various non-thiol antioxidants: dimethyl sulfoxide, dimetylthiourea, sodium salicylate, sodium formate, uric acid, catalase, and superoxide dismutase. Sodium Salicylate 173-190 heme oxygenase 1 Homo sapiens 62-67 15773552-4 2005 Further, to identify the ROS family members implicated in the HOX-1 induction, we also exposed cells to various non-thiol antioxidants: dimethyl sulfoxide, dimetylthiourea, sodium salicylate, sodium formate, uric acid, catalase, and superoxide dismutase. formic acid 192-206 heme oxygenase 1 Homo sapiens 62-67 15773552-4 2005 Further, to identify the ROS family members implicated in the HOX-1 induction, we also exposed cells to various non-thiol antioxidants: dimethyl sulfoxide, dimetylthiourea, sodium salicylate, sodium formate, uric acid, catalase, and superoxide dismutase. Uric Acid 208-217 heme oxygenase 1 Homo sapiens 62-67 15773552-5 2005 A partial inhibition of HOX-1 induction occurred in the presence of non-polar hydroxyl radical scavengers, dimethyl sulfoxide and dimetylthiourea. Hydroxyl Radical 78-94 heme oxygenase 1 Homo sapiens 24-29 15773552-5 2005 A partial inhibition of HOX-1 induction occurred in the presence of non-polar hydroxyl radical scavengers, dimethyl sulfoxide and dimetylthiourea. Dimethyl Sulfoxide 107-125 heme oxygenase 1 Homo sapiens 24-29 15773552-5 2005 A partial inhibition of HOX-1 induction occurred in the presence of non-polar hydroxyl radical scavengers, dimethyl sulfoxide and dimetylthiourea. dimetylthiourea 130-145 heme oxygenase 1 Homo sapiens 24-29 15773552-7 2005 Then, in order to determine, whether RNS scavenging is implicated in the HOX-1 down-regulation by thiol antioxidants, we took advantage of the capacity of suboptimal concentrations of the NO scavenger PTIO (2-phenyl-4,4,5,5-tetramethylimidazole-1-oxyl-3-oxide) to oxidize NO to nitrosating species. Sulfhydryl Compounds 98-103 heme oxygenase 1 Homo sapiens 73-78 15773552-3 2005 In this study we have examined, whether the scavenging of reactive oxygen and reactive nitrogen species (ROS and RNS) is the major cause for thiol-mediated suppression of the HOX-1 induction by NO. ros 105-108 heme oxygenase 1 Homo sapiens 175-180 15773552-3 2005 In this study we have examined, whether the scavenging of reactive oxygen and reactive nitrogen species (ROS and RNS) is the major cause for thiol-mediated suppression of the HOX-1 induction by NO. Sulfhydryl Compounds 141-146 heme oxygenase 1 Homo sapiens 175-180 15541371-0 2004 Rosuvastatin upregulates the antioxidant defense protein heme oxygenase-1. Rosuvastatin Calcium 0-12 heme oxygenase 1 Homo sapiens 57-73 15808651-5 2005 One of these genes that has been shown to mediate protective effects decodes the enzyme heme oxygenase-1 (HO-1), and an HO-1 downstream product, carbon monoxide (CO). Carbon Monoxide 145-160 heme oxygenase 1 Homo sapiens 120-124 15808651-5 2005 One of these genes that has been shown to mediate protective effects decodes the enzyme heme oxygenase-1 (HO-1), and an HO-1 downstream product, carbon monoxide (CO). Carbon Monoxide 162-164 heme oxygenase 1 Homo sapiens 106-110 15808651-5 2005 One of these genes that has been shown to mediate protective effects decodes the enzyme heme oxygenase-1 (HO-1), and an HO-1 downstream product, carbon monoxide (CO). Carbon Monoxide 162-164 heme oxygenase 1 Homo sapiens 120-124 15808653-5 2005 Donor animals were treated with cobalt protoporphyrin (CoPP; 5 mg/kg IP) 24 hours prior to organ harvesting to induce HO-1. cobaltiprotoporphyrin 32-53 heme oxygenase 1 Homo sapiens 118-122 15808653-5 2005 Donor animals were treated with cobalt protoporphyrin (CoPP; 5 mg/kg IP) 24 hours prior to organ harvesting to induce HO-1. cobaltiprotoporphyrin 55-59 heme oxygenase 1 Homo sapiens 118-122 15808653-6 2005 Controls remained untreated or received zinc protoporphyrin (ZnPP; 20 mg/kg, IP) to block HO-1 induction. zinc protoporphyrin 61-65 heme oxygenase 1 Homo sapiens 90-94 15541371-2 2004 This study explores the role of heme oxygenase-1 (HO-1) as target and potential mediator of rosuvastatin. Rosuvastatin Calcium 92-104 heme oxygenase 1 Homo sapiens 32-48 15541371-2 2004 This study explores the role of heme oxygenase-1 (HO-1) as target and potential mediator of rosuvastatin. Rosuvastatin Calcium 92-104 heme oxygenase 1 Homo sapiens 50-54 15541371-3 2004 In cultured human endothelial cells (ECV 304), rosuvastatin increased HO-1 mRNA and protein levels in a concentration-dependent fashion. Rosuvastatin Calcium 47-59 heme oxygenase 1 Homo sapiens 70-74 15541371-4 2004 HO-1 induction by rosuvastatin remained unaffected by mevalonate and N-nitro-L-arginine-methylester, showing that isoprenoid- and NO-dependent pathways were not involved. Rosuvastatin Calcium 18-30 heme oxygenase 1 Homo sapiens 0-4 15541371-6 2004 The HO-1 metabolite bilirubin, when added exogenously to the cells, virtually abolished NADPH-dependent oxidative stress. Bilirubin 20-29 heme oxygenase 1 Homo sapiens 4-8 15541371-6 2004 The HO-1 metabolite bilirubin, when added exogenously to the cells, virtually abolished NADPH-dependent oxidative stress. NADP 88-93 heme oxygenase 1 Homo sapiens 4-8 15541371-8 2004 Our results demonstrate that HO-1 is a target site and antioxidant mediator of rosuvastatin in endothelial cells. Rosuvastatin Calcium 79-91 heme oxygenase 1 Homo sapiens 29-33 15577407-6 2004 Treatment of cells with this protease inhibitor led to a significant increase in expression of the heme oxygenase-1 gene that could be reduced by 100 microM of the antioxidant ascorbate. Ascorbic Acid 176-185 heme oxygenase 1 Homo sapiens 99-115 15544924-3 2004 In cultured endothelial cells derived from human umbilical vein, simvastatin and lovastatin increased HO-1 mRNA levels in a concentration- and time-dependent fashion. Simvastatin 65-76 heme oxygenase 1 Homo sapiens 102-106 15544924-3 2004 In cultured endothelial cells derived from human umbilical vein, simvastatin and lovastatin increased HO-1 mRNA levels in a concentration- and time-dependent fashion. Lovastatin 81-91 heme oxygenase 1 Homo sapiens 102-106 15544924-5 2004 HO-1 mRNA induction was abrogated in the presence of actinomycin D and cycloheximide. Dactinomycin 53-66 heme oxygenase 1 Homo sapiens 0-4 15544924-5 2004 HO-1 mRNA induction was abrogated in the presence of actinomycin D and cycloheximide. Cycloheximide 71-84 heme oxygenase 1 Homo sapiens 0-4 15542064-0 2004 Diallyl sulfide induces heme oxygenase-1 through MAPK pathway. allyl sulfide 0-15 heme oxygenase 1 Homo sapiens 24-40 15542064-3 2004 In this study, we found that DAS can induce the expression of heme oxygenase-1 (HO-1), which plays a critical role in the cell defense system against oxidative stress. allyl sulfide 29-32 heme oxygenase 1 Homo sapiens 62-78 15542064-3 2004 In this study, we found that DAS can induce the expression of heme oxygenase-1 (HO-1), which plays a critical role in the cell defense system against oxidative stress. allyl sulfide 29-32 heme oxygenase 1 Homo sapiens 80-84 15542064-10 2004 N-Acetyl-cysteine blocked this increase of ROS production as well as DAS-induced ERK activation, Nrf2 protein expression and nuclear translocation, and ho-1 gene activation. Acetylcysteine 0-17 heme oxygenase 1 Homo sapiens 152-156 15542064-11 2004 The increase in HO-1 produced by DAS protected the HepG2 cells against toxicity by hydrogen peroxide or arachidonic acid. Hydrogen Peroxide 83-100 heme oxygenase 1 Homo sapiens 16-20 15542064-11 2004 The increase in HO-1 produced by DAS protected the HepG2 cells against toxicity by hydrogen peroxide or arachidonic acid. Arachidonic Acid 104-120 heme oxygenase 1 Homo sapiens 16-20 15542064-12 2004 These results suggest that DAS induces ho-1 through production of ROS, and Nrf2 and MAPK (ERK and p38) mediate this induction. allyl sulfide 27-30 heme oxygenase 1 Homo sapiens 39-43 15542064-12 2004 These results suggest that DAS induces ho-1 through production of ROS, and Nrf2 and MAPK (ERK and p38) mediate this induction. Reactive Oxygen Species 66-69 heme oxygenase 1 Homo sapiens 39-43 15542064-13 2004 Induction of ho-1 may play a role in the protective effects of DAS. allyl sulfide 63-66 heme oxygenase 1 Homo sapiens 13-17 15659834-0 2004 Effects of cyclopentenone prostaglandins on the expression of heme oxygenase-1 in MCF-7 cells. cyclopentenone 11-25 heme oxygenase 1 Homo sapiens 62-78 15465821-1 2004 Heme oxygenase-1 is an antioxidant defense enzyme that converts heme to biliverdin, iron, and carbon monoxide. Heme 64-68 heme oxygenase 1 Homo sapiens 0-16 15465821-1 2004 Heme oxygenase-1 is an antioxidant defense enzyme that converts heme to biliverdin, iron, and carbon monoxide. Biliverdine 72-82 heme oxygenase 1 Homo sapiens 0-16 15465821-1 2004 Heme oxygenase-1 is an antioxidant defense enzyme that converts heme to biliverdin, iron, and carbon monoxide. Iron 84-88 heme oxygenase 1 Homo sapiens 0-16 15465821-1 2004 Heme oxygenase-1 is an antioxidant defense enzyme that converts heme to biliverdin, iron, and carbon monoxide. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 0-16 15465821-7 2004 The effect of increasing concentrations of heme to up-regulate levels of heme oxygenase-1 was more pronounced when Bach-1 siRNA was present. Heme 43-47 heme oxygenase 1 Homo sapiens 73-89 15581622-5 2004 Isotopic 32P-labeling of HEK293T cells confirmed that HO-1 is a phosphoprotein and that the basal HO-1 phosphorylation is increased by Akt1 activation. Phosphorus-32 9-12 heme oxygenase 1 Homo sapiens 54-58 15581622-5 2004 Isotopic 32P-labeling of HEK293T cells confirmed that HO-1 is a phosphoprotein and that the basal HO-1 phosphorylation is increased by Akt1 activation. Phosphorus-32 9-12 heme oxygenase 1 Homo sapiens 98-102 15308469-4 2004 After it enters the cell, heme is degraded by heme oxygenase-1 (HO-1), and iron is released. Heme 26-30 heme oxygenase 1 Homo sapiens 46-62 15564944-2 2004 The authors hypothesized that propofol, because of its particular chemical structure, mitigates the effects of peroxynitrite-mediated oxidative stress and apoptosis by the induction of heme oxygenase (HO)-1 in primary cultured astroglial cells. Propofol 30-38 heme oxygenase 1 Homo sapiens 185-206 15564944-2 2004 The authors hypothesized that propofol, because of its particular chemical structure, mitigates the effects of peroxynitrite-mediated oxidative stress and apoptosis by the induction of heme oxygenase (HO)-1 in primary cultured astroglial cells. Peroxynitrous Acid 111-124 heme oxygenase 1 Homo sapiens 185-206 15659834-0 2004 Effects of cyclopentenone prostaglandins on the expression of heme oxygenase-1 in MCF-7 cells. Prostaglandins 26-40 heme oxygenase 1 Homo sapiens 62-78 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Heme 72-76 heme oxygenase 1 Homo sapiens 90-94 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Iron 185-189 heme oxygenase 1 Homo sapiens 72-88 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Iron 185-189 heme oxygenase 1 Homo sapiens 90-94 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Carbon Monoxide 191-206 heme oxygenase 1 Homo sapiens 72-88 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Carbon Monoxide 191-206 heme oxygenase 1 Homo sapiens 90-94 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Biliverdine 212-222 heme oxygenase 1 Homo sapiens 72-88 15659834-5 2004 In this study, we investigated the effect of cyPGs on the expression of heme oxygenase-1 (HO-1), a ubiquitous stress-responsive enzyme that catalyzes oxidative cleavage of heme to form iron, carbon monoxide, and biliverdin. Biliverdine 212-222 heme oxygenase 1 Homo sapiens 90-94 15659834-6 2004 Treatment of the human breast cancer cell line (MCF-7) with 15d-PGJ(2) resulted in a concentration- and time-dependent increase in the expression of HO-1, whereas prostaglandin A(2) (PGA(2)) and the non-PG derivative 2-cyclopenten-1-one failed to induce HO-1 expression at the protein level. Prostaglandins 64-66 heme oxygenase 1 Homo sapiens 149-153 15659834-6 2004 Treatment of the human breast cancer cell line (MCF-7) with 15d-PGJ(2) resulted in a concentration- and time-dependent increase in the expression of HO-1, whereas prostaglandin A(2) (PGA(2)) and the non-PG derivative 2-cyclopenten-1-one failed to induce HO-1 expression at the protein level. Prostaglandins 64-66 heme oxygenase 1 Homo sapiens 254-258 15659834-6 2004 Treatment of the human breast cancer cell line (MCF-7) with 15d-PGJ(2) resulted in a concentration- and time-dependent increase in the expression of HO-1, whereas prostaglandin A(2) (PGA(2)) and the non-PG derivative 2-cyclopenten-1-one failed to induce HO-1 expression at the protein level. cyclopentenone 217-236 heme oxygenase 1 Homo sapiens 149-153 15659834-11 2004 Thus, the induction of HO-1 expression and the activation of Akt/PKB by 15d-PGJ(2) and PGA(2) are likely to confer cytoprotective or antiapoptotic effects exerted by these cyPGs. 15d-pgj 72-79 heme oxygenase 1 Homo sapiens 23-27 15659834-11 2004 Thus, the induction of HO-1 expression and the activation of Akt/PKB by 15d-PGJ(2) and PGA(2) are likely to confer cytoprotective or antiapoptotic effects exerted by these cyPGs. prostaglandin A2 87-93 heme oxygenase 1 Homo sapiens 23-27 15504750-0 2004 The HIV protease inhibitor ritonavir synergizes with butyrate for induction of apoptotic cell death and mediates expression of heme oxygenase-1 in DLD-1 colon carcinoma cells. Ritonavir 27-36 heme oxygenase 1 Homo sapiens 127-143 15560792-5 2004 PCC 6803 (Syn HO-1), in complex with heme at 2.5 A resolution. Heme 37-41 heme oxygenase 1 Homo sapiens 14-18 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. Ritonavir 0-9 heme oxygenase 1 Homo sapiens 18-22 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. SB 203580 49-57 heme oxygenase 1 Homo sapiens 18-22 15504750-9 2004 Ritonavir-induced HO-1 protein was suppressed by SB203580 or SB202190 and preceded by immediate upregulation of cellular c-Fos and c-Jun protein levels. 4-(4-fluorophenyl)-2-(4-hydroxyphenyl)-5-(4-pyridyl)imidazole 61-69 heme oxygenase 1 Homo sapiens 18-22 15504750-11 2004 The present data suggest that ritonavir has the potential to curb colon carcinogenesis by reducing cell growth via mechanisms that include apoptosis and by simultaneously modulating colonic inflammation via induction of anti-inflammatory HO-1. Ritonavir 30-39 heme oxygenase 1 Homo sapiens 238-242 15642322-9 2004 Analysis with real-time PCR showed a maximum 3-6-fold increase in mRNA levels 9 hr after the 3 hr O2-exposure for the enzymes heme oxygenase-1 (HO-1), MnSOD and TrxR1 (the cytoplasmic form of TrxR). Oxygen 98-100 heme oxygenase 1 Homo sapiens 126-142 15642322-9 2004 Analysis with real-time PCR showed a maximum 3-6-fold increase in mRNA levels 9 hr after the 3 hr O2-exposure for the enzymes heme oxygenase-1 (HO-1), MnSOD and TrxR1 (the cytoplasmic form of TrxR). Oxygen 98-100 heme oxygenase 1 Homo sapiens 144-148 15642322-11 2004 Initial upregulation of message for HO-1 occurred a few hours before any upregulation of MnSOD could be detected, suggesting that release of free iron from the degradation of heme by HO-1 may have played a role in the upregulation of the dismutase. Iron 146-150 heme oxygenase 1 Homo sapiens 36-40 15642322-11 2004 Initial upregulation of message for HO-1 occurred a few hours before any upregulation of MnSOD could be detected, suggesting that release of free iron from the degradation of heme by HO-1 may have played a role in the upregulation of the dismutase. Iron 146-150 heme oxygenase 1 Homo sapiens 183-187 15547665-3 2004 Heme oxygenase-1 (HO-1) is known to be induced not only by its substrate, heme, but also by various oxidative stresses, and thought to play an important role in the protection of the host from oxidative tissue injuries. Heme 74-78 heme oxygenase 1 Homo sapiens 0-16 15547665-3 2004 Heme oxygenase-1 (HO-1) is known to be induced not only by its substrate, heme, but also by various oxidative stresses, and thought to play an important role in the protection of the host from oxidative tissue injuries. Heme 74-78 heme oxygenase 1 Homo sapiens 18-22 15547665-8 2004 These findings suggest that, in the liver of ALF patients, there may be an increase in free heme concentration which up-regulates HO-1 gene expression, while down-regulating ALAS1 gene expression, resulting in markedly altered heme metabolism and liver function. Heme 92-96 heme oxygenase 1 Homo sapiens 130-134 15642322-11 2004 Initial upregulation of message for HO-1 occurred a few hours before any upregulation of MnSOD could be detected, suggesting that release of free iron from the degradation of heme by HO-1 may have played a role in the upregulation of the dismutase. Heme 175-179 heme oxygenase 1 Homo sapiens 36-40 15642322-11 2004 Initial upregulation of message for HO-1 occurred a few hours before any upregulation of MnSOD could be detected, suggesting that release of free iron from the degradation of heme by HO-1 may have played a role in the upregulation of the dismutase. Heme 175-179 heme oxygenase 1 Homo sapiens 183-187 15531134-0 2004 Effects of 1alpha,25 dihydroxyvitamin D3 on the expression of HO-1 and GFAP in glial cells of the photothrombotically lesioned cerebral cortex. Calcitriol 11-40 heme oxygenase 1 Homo sapiens 62-66 15531134-3 2004 The inducible enzyme, heme oxygenase-1 metabolizes and thus detoxifies free heme to the powerful endogenous antioxidants biliverdin and bilirubin therefore enhancing neuroprotection. Biliverdine 121-131 heme oxygenase 1 Homo sapiens 22-38 15531134-3 2004 The inducible enzyme, heme oxygenase-1 metabolizes and thus detoxifies free heme to the powerful endogenous antioxidants biliverdin and bilirubin therefore enhancing neuroprotection. Bilirubin 136-145 heme oxygenase 1 Homo sapiens 22-38 15531134-5 2004 We studied the effects of 1,25-D3 treatment on heme oxygenase-1 expression following focal cortical ischemia elicited by photothrombosis. Calcitriol 26-33 heme oxygenase 1 Homo sapiens 47-63 15531134-6 2004 Postlesional treatment with 1,25-D3 (4 microg/kg body weight) resulted in a transient, but significant upregulation of glial heme oxygenase-1 immunoreactivity concomitant with a reduction in glial fibrillary acidic protein immunoreactivity in remote cortical regions affected by a secondary spread of injury, whereas the size of the lesion"s core remained unaffected. Calcitriol 28-35 heme oxygenase 1 Homo sapiens 125-141 15450936-5 2004 Total GSH level decreased markedly (50% of control) by 2 h, began to recover at 4 h, returned to control level by 6 h and increased above the control level during 10-24 h. Collectively, these results indicated that overproduced O2*- depletes GSH and triggers induction of xCT, HO-1, iNOS and HO-1 expression in sequence. Glutathione 6-9 heme oxygenase 1 Homo sapiens 277-281 15450936-5 2004 Total GSH level decreased markedly (50% of control) by 2 h, began to recover at 4 h, returned to control level by 6 h and increased above the control level during 10-24 h. Collectively, these results indicated that overproduced O2*- depletes GSH and triggers induction of xCT, HO-1, iNOS and HO-1 expression in sequence. Glutathione 6-9 heme oxygenase 1 Homo sapiens 292-296 15450936-5 2004 Total GSH level decreased markedly (50% of control) by 2 h, began to recover at 4 h, returned to control level by 6 h and increased above the control level during 10-24 h. Collectively, these results indicated that overproduced O2*- depletes GSH and triggers induction of xCT, HO-1, iNOS and HO-1 expression in sequence. Oxygen 228-230 heme oxygenase 1 Homo sapiens 277-281 15450936-5 2004 Total GSH level decreased markedly (50% of control) by 2 h, began to recover at 4 h, returned to control level by 6 h and increased above the control level during 10-24 h. Collectively, these results indicated that overproduced O2*- depletes GSH and triggers induction of xCT, HO-1, iNOS and HO-1 expression in sequence. Oxygen 228-230 heme oxygenase 1 Homo sapiens 292-296 15450936-7 2004 When this iNOS-derived delivery of NO* was combined with prior depletion of GSH using buthioninesulfoximine, an inhibitor of GSH biosynthesis, induction of HO-1 was potentiated. Glutathione 76-79 heme oxygenase 1 Homo sapiens 156-160 15450936-7 2004 When this iNOS-derived delivery of NO* was combined with prior depletion of GSH using buthioninesulfoximine, an inhibitor of GSH biosynthesis, induction of HO-1 was potentiated. Buthionine Sulfoximine 86-107 heme oxygenase 1 Homo sapiens 156-160 15450936-7 2004 When this iNOS-derived delivery of NO* was combined with prior depletion of GSH using buthioninesulfoximine, an inhibitor of GSH biosynthesis, induction of HO-1 was potentiated. Glutathione 125-128 heme oxygenase 1 Homo sapiens 156-160 15571180-5 2004 PATIENTS AND METHODS: To this end, the gene expression of p22phox, a NAD(P)H oxidase subunit closely linked with the generation of superoxide anions and of Heme oxygenase-1 (HO-1), induced by and protective from oxidative stress, were evaluated by RT-PCR in mononuclear cells from 5 patients under 3 times a week chronic bicarbonate dialysis. Bicarbonates 321-332 heme oxygenase 1 Homo sapiens 156-172 15560792-7 2004 Although the heme pocket of heme-Syn HO-1 is, for the most part, similar to that of mammalian HO-1, they differ in such features as the flexibility of the distal helix and hydrophobicity. Heme 13-17 heme oxygenase 1 Homo sapiens 37-41 15560792-7 2004 Although the heme pocket of heme-Syn HO-1 is, for the most part, similar to that of mammalian HO-1, they differ in such features as the flexibility of the distal helix and hydrophobicity. Heme 28-32 heme oxygenase 1 Homo sapiens 37-41 15560792-7 2004 Although the heme pocket of heme-Syn HO-1 is, for the most part, similar to that of mammalian HO-1, they differ in such features as the flexibility of the distal helix and hydrophobicity. Heme 28-32 heme oxygenase 1 Homo sapiens 94-98 15560792-10 2004 The surfaces of the heme binding sides are both positively charged, but this patch of Syn HO-1 is narrow compared to that of mammalian HO-1. Heme 20-24 heme oxygenase 1 Homo sapiens 90-94 15560792-10 2004 The surfaces of the heme binding sides are both positively charged, but this patch of Syn HO-1 is narrow compared to that of mammalian HO-1. Heme 20-24 heme oxygenase 1 Homo sapiens 135-139 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Iron 92-96 heme oxygenase 1 Homo sapiens 28-32 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Iron 92-96 heme oxygenase 1 Homo sapiens 155-159 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Sulfur 97-103 heme oxygenase 1 Homo sapiens 28-32 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Sulfur 97-103 heme oxygenase 1 Homo sapiens 155-159 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Heme 19-23 heme oxygenase 1 Homo sapiens 28-32 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Heme 19-23 heme oxygenase 1 Homo sapiens 155-159 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Iron 138-142 heme oxygenase 1 Homo sapiens 28-32 15560792-12 2004 A docking model of heme-Syn HO-1 and ferredoxin suggests indirect electron transfer from an iron-sulfur cluster in ferredoxin to the heme iron of heme-Syn HO-1. Iron 138-142 heme oxygenase 1 Homo sapiens 155-159 15451438-0 2004 Augmented heme oxygenase-1 induces prostaglandin uptake via the prostaglandin transporter in micro-vascular endothelial cells. Prostaglandins 35-48 heme oxygenase 1 Homo sapiens 10-26 15591777-4 2004 Scavenging carbon monoxide (CO), one of the by-products of heme degradation by HO-1, significantly attenuated HO-1-mediated suppression of iNOS gene induction as revealed by Northern blot analysis. Carbon Monoxide 11-26 heme oxygenase 1 Homo sapiens 79-83 15591777-4 2004 Scavenging carbon monoxide (CO), one of the by-products of heme degradation by HO-1, significantly attenuated HO-1-mediated suppression of iNOS gene induction as revealed by Northern blot analysis. Carbon Monoxide 11-26 heme oxygenase 1 Homo sapiens 110-114 15591777-4 2004 Scavenging carbon monoxide (CO), one of the by-products of heme degradation by HO-1, significantly attenuated HO-1-mediated suppression of iNOS gene induction as revealed by Northern blot analysis. Carbon Monoxide 28-30 heme oxygenase 1 Homo sapiens 79-83 15591777-4 2004 Scavenging carbon monoxide (CO), one of the by-products of heme degradation by HO-1, significantly attenuated HO-1-mediated suppression of iNOS gene induction as revealed by Northern blot analysis. Carbon Monoxide 28-30 heme oxygenase 1 Homo sapiens 110-114 15591777-9 2004 CO appears to mediate the suppressive effect of HO-1, at least in part, through downregulating transcriptional activation of the iNOS gene via a cGMP-dependent pathway. Cyclic GMP 145-149 heme oxygenase 1 Homo sapiens 48-52 15519649-5 2004 HO-1 enzyme activity was highest during warm hypoxia, followed by cold hypoxia and CM which was confirmed by intracellular Fe(2+) formation. ammonium ferrous sulfate 123-129 heme oxygenase 1 Homo sapiens 0-4 15474356-9 2004 These results support the hypothesis that HO-1 protects astrocytes from heme-mediated oxidative injury. Heme 72-76 heme oxygenase 1 Homo sapiens 42-46 15530192-12 2004 Bilirubin, but not carboxyhemoglobin, correlated with heme oxygenase-1 expression (p = .0013). Bilirubin 0-9 heme oxygenase 1 Homo sapiens 54-70 15530192-14 2004 Heme oxygenase-1 expression correlated with serum bilirubin levels. Bilirubin 50-59 heme oxygenase 1 Homo sapiens 0-16 15530192-15 2004 The increase in heme oxygenase-1 expression may add to the understanding of the increase in serum bilirubin observed in patients with SIRS/sepsis. Bilirubin 98-107 heme oxygenase 1 Homo sapiens 16-32 15522396-0 2004 Crystal structures of ferrous and ferrous-NO forms of verdoheme in a complex with human heme oxygenase-1: catalytic implications for heme cleavage. ammonium ferrous sulfate 22-29 heme oxygenase 1 Homo sapiens 88-104 15522396-0 2004 Crystal structures of ferrous and ferrous-NO forms of verdoheme in a complex with human heme oxygenase-1: catalytic implications for heme cleavage. ammonium ferrous sulfate 34-41 heme oxygenase 1 Homo sapiens 88-104 15522396-0 2004 Crystal structures of ferrous and ferrous-NO forms of verdoheme in a complex with human heme oxygenase-1: catalytic implications for heme cleavage. Heme 59-63 heme oxygenase 1 Homo sapiens 88-104 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. mesophorphyrin 163-177 heme oxygenase 1 Homo sapiens 145-149 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. mesophorphyrin 163-177 heme oxygenase 1 Homo sapiens 145-149 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. Prostaglandins 98-100 heme oxygenase 1 Homo sapiens 57-61 15451438-1 2004 Previous studies show that expression of heme oxygenase-1 (HO-1) in endothelial cells results in decreased cyclooxygenase expression and prostaglandin (PG) levels through limiting heme availability. Prostaglandins 137-150 heme oxygenase 1 Homo sapiens 41-57 15451438-7 2004 Therefore, PG clearance via PGT may contribute to the cellular regulation of PG levels by HO-1. Prostaglandins 11-13 heme oxygenase 1 Homo sapiens 90-94 15451438-1 2004 Previous studies show that expression of heme oxygenase-1 (HO-1) in endothelial cells results in decreased cyclooxygenase expression and prostaglandin (PG) levels through limiting heme availability. Prostaglandins 137-150 heme oxygenase 1 Homo sapiens 59-63 15451438-1 2004 Previous studies show that expression of heme oxygenase-1 (HO-1) in endothelial cells results in decreased cyclooxygenase expression and prostaglandin (PG) levels through limiting heme availability. Prostaglandins 152-154 heme oxygenase 1 Homo sapiens 41-57 15451438-7 2004 Therefore, PG clearance via PGT may contribute to the cellular regulation of PG levels by HO-1. pgt 28-31 heme oxygenase 1 Homo sapiens 90-94 15451438-1 2004 Previous studies show that expression of heme oxygenase-1 (HO-1) in endothelial cells results in decreased cyclooxygenase expression and prostaglandin (PG) levels through limiting heme availability. Prostaglandins 152-154 heme oxygenase 1 Homo sapiens 59-63 15451438-7 2004 Therefore, PG clearance via PGT may contribute to the cellular regulation of PG levels by HO-1. Prostaglandins 28-30 heme oxygenase 1 Homo sapiens 90-94 15451438-1 2004 Previous studies show that expression of heme oxygenase-1 (HO-1) in endothelial cells results in decreased cyclooxygenase expression and prostaglandin (PG) levels through limiting heme availability. Heme 41-45 heme oxygenase 1 Homo sapiens 59-63 15451438-2 2004 Regulation of PGs, important inflammatory mediators, may contribute to the anti-inflammatory potential of HO-1. Phosphatidylglycerols 14-17 heme oxygenase 1 Homo sapiens 106-110 15451438-3 2004 Here we examine the effects of HO-1 expression on PG clearance via the prostaglandin transporter (PGT). Prostaglandins 50-52 heme oxygenase 1 Homo sapiens 31-35 15451438-4 2004 Endothelial cells expressing human HO-1 via retroviral transfer exhibit approximately 7-fold higher levels of PGT RNA and equivalently elevated uptake of [(3)H]PGE(2). Prostaglandins E 160-163 heme oxygenase 1 Homo sapiens 35-39 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. stannous chloride 24-41 heme oxygenase 1 Homo sapiens 57-61 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. stannous chloride 24-41 heme oxygenase 1 Homo sapiens 145-149 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. stannous chloride 24-41 heme oxygenase 1 Homo sapiens 145-149 15451438-6 2004 Treatment of cells with stannous chloride, an inducer of HO-1, results in increased expression of PGT while incubation of cells expressing human HO-1 with stannic mesophorphyrin, a substrate inhibitor of HO-1, decreases PG uptake. pgt 98-101 heme oxygenase 1 Homo sapiens 57-61 15242350-2 2004 HO-1 transcription is induced by a vast array of compounds including, but certainly not limited to, haem and heavy metals such as cadmium. Cadmium 130-137 heme oxygenase 1 Homo sapiens 0-4 15242350-4 2004 Dimethylsulphate in vivo footprinting studies have identified three protected guanine residues in the E-box of the HO-1 proximal promoter. dimethyl sulfate 0-16 heme oxygenase 1 Homo sapiens 115-119 15242350-4 2004 Dimethylsulphate in vivo footprinting studies have identified three protected guanine residues in the E-box of the HO-1 proximal promoter. Guanine 78-85 heme oxygenase 1 Homo sapiens 115-119 15242350-8 2004 These results demonstrate for the first time that USF proteins bind to the human HO-1 promoter in vivo and are required for high-level expression of HO-1 by haem and cadmium in human renal epithelial cells. Heme 157-161 heme oxygenase 1 Homo sapiens 81-85 15242350-8 2004 These results demonstrate for the first time that USF proteins bind to the human HO-1 promoter in vivo and are required for high-level expression of HO-1 by haem and cadmium in human renal epithelial cells. Heme 157-161 heme oxygenase 1 Homo sapiens 149-153 15242350-8 2004 These results demonstrate for the first time that USF proteins bind to the human HO-1 promoter in vivo and are required for high-level expression of HO-1 by haem and cadmium in human renal epithelial cells. Cadmium 166-173 heme oxygenase 1 Homo sapiens 81-85 15242350-8 2004 These results demonstrate for the first time that USF proteins bind to the human HO-1 promoter in vivo and are required for high-level expression of HO-1 by haem and cadmium in human renal epithelial cells. Cadmium 166-173 heme oxygenase 1 Homo sapiens 149-153 15378604-4 2004 Effect of HO-1 production on the cell injury induced by hydrogen peroxide was evaluated after hemin stimulation and after HO-1 gene transfection. Hydrogen Peroxide 56-73 heme oxygenase 1 Homo sapiens 10-14 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Oxygen 45-47 heme oxygenase 1 Homo sapiens 6-22 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Oxygen 45-47 heme oxygenase 1 Homo sapiens 24-29 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Heme 6-10 heme oxygenase 1 Homo sapiens 24-29 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 79-89 heme oxygenase 1 Homo sapiens 6-22 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 79-89 heme oxygenase 1 Homo sapiens 24-29 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 91-93 heme oxygenase 1 Homo sapiens 6-22 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 91-93 heme oxygenase 1 Homo sapiens 24-29 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 104-108 heme oxygenase 1 Homo sapiens 6-22 15297453-1 2004 Human heme oxygenase-1 (hHO-1) catalyzes the O2-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 104-108 heme oxygenase 1 Homo sapiens 24-29 15297453-6 2004 We report here that hHO-1 cleaves 5-phenylheme to biliverdin IXalpha and oxidizes 15-phenylheme at the alpha-meso position to give 10-phenylbiliverdin IXalpha. 5-phenylheme 34-46 heme oxygenase 1 Homo sapiens 20-25 15297453-6 2004 We report here that hHO-1 cleaves 5-phenylheme to biliverdin IXalpha and oxidizes 15-phenylheme at the alpha-meso position to give 10-phenylbiliverdin IXalpha. Biliverdine 50-68 heme oxygenase 1 Homo sapiens 20-25 15297453-6 2004 We report here that hHO-1 cleaves 5-phenylheme to biliverdin IXalpha and oxidizes 15-phenylheme at the alpha-meso position to give 10-phenylbiliverdin IXalpha. 15-phenylheme 82-95 heme oxygenase 1 Homo sapiens 20-25 15297453-6 2004 We report here that hHO-1 cleaves 5-phenylheme to biliverdin IXalpha and oxidizes 15-phenylheme at the alpha-meso position to give 10-phenylbiliverdin IXalpha. 10-phenylbiliverdin ixalpha 131-158 heme oxygenase 1 Homo sapiens 20-25 15297453-8 2004 The 2.29- and 2.11-A crystal structures of the hHO-1 complexes with 1- and 15-phenylheme, respectively, show clear electron density for both the 5- and 15-phenyl rings in both molecules of the asymmetric unit. phenylheme 78-88 heme oxygenase 1 Homo sapiens 47-52 15297453-9 2004 The overall structure of 15-phenylheme-hHO-1 is similar to that of heme-hHO-1 except for small changes in distal residues 141-150 and in the proximal Lys18 and Lys22. octadeca(lysine) 150-155 heme oxygenase 1 Homo sapiens 39-44 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. phenyl-substituted heme 41-64 heme oxygenase 1 Homo sapiens 20-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. phenyl-substituted heme 41-64 heme oxygenase 1 Homo sapiens 21-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. Heme 15-19 heme oxygenase 1 Homo sapiens 20-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. Heme 15-19 heme oxygenase 1 Homo sapiens 21-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. meso 251-255 heme oxygenase 1 Homo sapiens 20-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. meso 251-255 heme oxygenase 1 Homo sapiens 21-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. Carbon 256-262 heme oxygenase 1 Homo sapiens 20-25 15297453-10 2004 In the 5-phenylheme-hHO-1 structure, the phenyl-substituted heme occupies the same position as heme in the heme-HO-1 complex but the 5-phenyl substituent disrupts the rigid hydrophobic wall of residues Met34, Phe214, and residues 26-42 near the alpha-meso carbon. Carbon 256-262 heme oxygenase 1 Homo sapiens 21-25 15345146-3 2004 Moreover, heme oxygenase-1 (HO-1), a ubiquitous stress-inducible enzyme that is induced by ROS and NO, was recently discovered to be involved in angiogenesis. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 10-26 15345141-4 2004 Free heme, i.e., a protein-unbound heme, exists in cells at a very minute concentration and exerts regulatory functions such as the repression of nonspecific delta-aminolevulinate synthase expression and the induction of microsomal heme oxygenase-1 (HO-1). Heme 5-9 heme oxygenase 1 Homo sapiens 232-248 15345146-3 2004 Moreover, heme oxygenase-1 (HO-1), a ubiquitous stress-inducible enzyme that is induced by ROS and NO, was recently discovered to be involved in angiogenesis. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 28-32 15345141-4 2004 Free heme, i.e., a protein-unbound heme, exists in cells at a very minute concentration and exerts regulatory functions such as the repression of nonspecific delta-aminolevulinate synthase expression and the induction of microsomal heme oxygenase-1 (HO-1). Heme 5-9 heme oxygenase 1 Homo sapiens 250-254 15345141-4 2004 Free heme, i.e., a protein-unbound heme, exists in cells at a very minute concentration and exerts regulatory functions such as the repression of nonspecific delta-aminolevulinate synthase expression and the induction of microsomal heme oxygenase-1 (HO-1). Heme 35-39 heme oxygenase 1 Homo sapiens 232-248 15345147-2 2004 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 15345141-4 2004 Free heme, i.e., a protein-unbound heme, exists in cells at a very minute concentration and exerts regulatory functions such as the repression of nonspecific delta-aminolevulinate synthase expression and the induction of microsomal heme oxygenase-1 (HO-1). Heme 35-39 heme oxygenase 1 Homo sapiens 250-254 15345147-2 2004 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 15345141-5 2004 The latter gene expression occurs by way of free heme-mediated derepression of Bach1, a mammalian heme-responsive transcription factor that suppresses the activation of the HO-1 gene. Heme 49-53 heme oxygenase 1 Homo sapiens 173-177 15345143-3 2004 Free heme, an entity that can be generated by UVA irradiation of cells, also appears to be a critical intermediate that can directly influence both the transcriptional activation and repression of the HO-1 gene. Heme 5-9 heme oxygenase 1 Homo sapiens 201-205 15345147-2 2004 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. Heme 109-113 heme oxygenase 1 Homo sapiens 0-16 15345147-2 2004 Heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, is induced not only by its substrate, heme, but also by oxidative stress. Heme 109-113 heme oxygenase 1 Homo sapiens 18-22 15345147-3 2004 In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. Heme 148-152 heme oxygenase 1 Homo sapiens 65-69 15345147-3 2004 In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. Carbon Monoxide 280-295 heme oxygenase 1 Homo sapiens 65-69 15345147-3 2004 In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. Biliverdine 297-315 heme oxygenase 1 Homo sapiens 65-69 15345147-3 2004 In various models of oxidative tissue injuries, the induction of HO-1 confers protection on tissues from further damages by removing the prooxidant heme, or by virtue of the antioxidative, antiinflammatory, and/or antiapoptotic actions of one or more of the three products, i.e., carbon monoxide, biliverdin IXalpha, and iron by HO reaction. Iron 321-325 heme oxygenase 1 Homo sapiens 65-69 15345152-3 2004 As a further complication, heme and cadmium, two potent inducers of the hmox-1 gene, inhibit Bach1 function by different mechanisms-by inhibition of DNA binding or promotion of nuclear export, respectively. Heme 27-31 heme oxygenase 1 Homo sapiens 72-78 15345152-3 2004 As a further complication, heme and cadmium, two potent inducers of the hmox-1 gene, inhibit Bach1 function by different mechanisms-by inhibition of DNA binding or promotion of nuclear export, respectively. Cadmium 36-43 heme oxygenase 1 Homo sapiens 72-78 15602829-0 2004 Induction of heme oxygenase-1 in human hepatocytes to protect them from ethanol-induced cytotoxicity. Ethanol 72-79 heme oxygenase 1 Homo sapiens 13-29 15636365-1 2004 Human Heme Oxygenase-1 (hHO-1) is the rate-limiting enzyme in the catabolism reaction of heme, which directly regulates the concentration of bilirubin in human body. Heme 89-93 heme oxygenase 1 Homo sapiens 6-22 15636365-1 2004 Human Heme Oxygenase-1 (hHO-1) is the rate-limiting enzyme in the catabolism reaction of heme, which directly regulates the concentration of bilirubin in human body. Heme 89-93 heme oxygenase 1 Homo sapiens 24-29 15636365-1 2004 Human Heme Oxygenase-1 (hHO-1) is the rate-limiting enzyme in the catabolism reaction of heme, which directly regulates the concentration of bilirubin in human body. Bilirubin 141-150 heme oxygenase 1 Homo sapiens 6-22 15636365-1 2004 Human Heme Oxygenase-1 (hHO-1) is the rate-limiting enzyme in the catabolism reaction of heme, which directly regulates the concentration of bilirubin in human body. Bilirubin 141-150 heme oxygenase 1 Homo sapiens 24-29 15636365-5 2004 The concentration of hHO-1 in 30%-60% saturation (NH4)2SO4 components and in fractions through twice column chromatography was 3.6-fold and 30-fold higher than that in initial product, respectively. Ammonium Sulfate 49-58 heme oxygenase 1 Homo sapiens 21-26 15458743-0 2004 Heme oxygenase-1 mediates up-regulation of adhesion molecule expression induced by peroxynitrite in endothelial cells. Peroxynitrous Acid 83-96 heme oxygenase 1 Homo sapiens 0-16 15458743-3 2004 This study was to test whether EC oxidative stress induced by peroxynitrite could up-regulate EC CAM expression, and whether heme oxygenase-1 (HO-1) has protective effects on this peroxynitrite-induced cellular response. Peroxynitrous Acid 180-193 heme oxygenase 1 Homo sapiens 125-141 15458743-3 2004 This study was to test whether EC oxidative stress induced by peroxynitrite could up-regulate EC CAM expression, and whether heme oxygenase-1 (HO-1) has protective effects on this peroxynitrite-induced cellular response. Peroxynitrous Acid 180-193 heme oxygenase 1 Homo sapiens 143-147 15458743-13 2004 CONCLUSIONS: Peroxynitrite-induced EC oxidative stress produces differential effects on CAM expression, which may be mediated by HO-1 regulation. Peroxynitrous Acid 13-26 heme oxygenase 1 Homo sapiens 129-133 15337692-0 2004 Simvastatin induces heme oxygenase-1: a novel mechanism of vessel protection. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 20-36 15337692-3 2004 Here, we studied whether heme oxygenase-1 (HO-1), an important cytoprotective molecule, is induced by simvastatin and the role of HO-1 in the pleiotropic effects of simvastatin. Simvastatin 102-113 heme oxygenase 1 Homo sapiens 25-41 15337692-3 2004 Here, we studied whether heme oxygenase-1 (HO-1), an important cytoprotective molecule, is induced by simvastatin and the role of HO-1 in the pleiotropic effects of simvastatin. Simvastatin 102-113 heme oxygenase 1 Homo sapiens 43-47 15337692-5 2004 The induction of HO-1 by simvastatin was not found in cultured endothelial cells and macrophages. Simvastatin 25-36 heme oxygenase 1 Homo sapiens 17-21 15337692-7 2004 Treating VSMCs with zinc protoporphyrin, an HO-1 inhibitor, or HO-1 small interfering RNA (siRNA) blocked the antiinflammatory effect of simvastatin, including the inhibition of nuclear factor-kappaB activation and nitric oxide production. Simvastatin 137-148 heme oxygenase 1 Homo sapiens 44-48 15337692-7 2004 Treating VSMCs with zinc protoporphyrin, an HO-1 inhibitor, or HO-1 small interfering RNA (siRNA) blocked the antiinflammatory effect of simvastatin, including the inhibition of nuclear factor-kappaB activation and nitric oxide production. Simvastatin 137-148 heme oxygenase 1 Homo sapiens 63-67 15337692-7 2004 Treating VSMCs with zinc protoporphyrin, an HO-1 inhibitor, or HO-1 small interfering RNA (siRNA) blocked the antiinflammatory effect of simvastatin, including the inhibition of nuclear factor-kappaB activation and nitric oxide production. Nitric Oxide 215-227 heme oxygenase 1 Homo sapiens 63-67 15337692-8 2004 Blockade of HO-1 also abolished the simvastatin-induced p21(Waf1) and the associated antiproliferative effect. Simvastatin 36-47 heme oxygenase 1 Homo sapiens 12-16 15337692-9 2004 Simvastatin activated p38 and Akt in VSMCs, and the respective inhibitors of p38 and phosphoinositide 3-kinase (PI3K) greatly reduced the level of simvastatin-induced HO-1, which suggests the involvement of p38 and the PI3K-Akt pathway in HO-1 induction. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 167-171 15337692-9 2004 Simvastatin activated p38 and Akt in VSMCs, and the respective inhibitors of p38 and phosphoinositide 3-kinase (PI3K) greatly reduced the level of simvastatin-induced HO-1, which suggests the involvement of p38 and the PI3K-Akt pathway in HO-1 induction. Simvastatin 0-11 heme oxygenase 1 Homo sapiens 239-243 15337692-9 2004 Simvastatin activated p38 and Akt in VSMCs, and the respective inhibitors of p38 and phosphoinositide 3-kinase (PI3K) greatly reduced the level of simvastatin-induced HO-1, which suggests the involvement of p38 and the PI3K-Akt pathway in HO-1 induction. Simvastatin 147-158 heme oxygenase 1 Homo sapiens 167-171 15337692-9 2004 Simvastatin activated p38 and Akt in VSMCs, and the respective inhibitors of p38 and phosphoinositide 3-kinase (PI3K) greatly reduced the level of simvastatin-induced HO-1, which suggests the involvement of p38 and the PI3K-Akt pathway in HO-1 induction. Simvastatin 147-158 heme oxygenase 1 Homo sapiens 239-243 15337692-10 2004 CONCLUSIONS: Simvastatin activates HO-1 in VSMCs in vitro and in vivo. Simvastatin 13-24 heme oxygenase 1 Homo sapiens 35-39 15337692-11 2004 The antiinflammatory and antiproliferative effects of simvastatin occur largely through the induced HO-1. Simvastatin 54-65 heme oxygenase 1 Homo sapiens 100-104 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. ammonium ferrous sulfate 506-510 heme oxygenase 1 Homo sapiens 49-53 15602829-9 2004 Furthermore, we demonstrated that pre-administration of cobalt protoporphyrin (CoPP) induced HO-1 in human hepatocytes, and prevented an increase of MDA and a decrease of GSH. cobaltiprotoporphyrin 56-77 heme oxygenase 1 Homo sapiens 93-97 15602829-9 2004 Furthermore, we demonstrated that pre-administration of cobalt protoporphyrin (CoPP) induced HO-1 in human hepatocytes, and prevented an increase of MDA and a decrease of GSH. cobaltiprotoporphyrin 79-83 heme oxygenase 1 Homo sapiens 93-97 15602829-10 2004 These effects could be partially reversed by zinc protoporphyrin (ZnPP), an antagonist of HO-1 induction. zinc protoporphyrin 45-64 heme oxygenase 1 Homo sapiens 90-94 15602829-10 2004 These effects could be partially reversed by zinc protoporphyrin (ZnPP), an antagonist of HO-1 induction. zinc protoporphyrin 66-70 heme oxygenase 1 Homo sapiens 90-94 15602829-11 2004 CONCLUSION: HO-1 expression in cells or organs could lead to new strategies for better prevention and treatment of ethanol-induced oxidative damage in human liver. Ethanol 115-122 heme oxygenase 1 Homo sapiens 12-16 15602829-1 2004 UNLABELLED: We investigated the relationship between ethanol exposure and heme oxygenase (HO-1) in human hepatocytes in order to ascertain if induction of HO-1 can prevent ethanol induced cellular damage. Ethanol 53-60 heme oxygenase 1 Homo sapiens 90-94 15602829-1 2004 UNLABELLED: We investigated the relationship between ethanol exposure and heme oxygenase (HO-1) in human hepatocytes in order to ascertain if induction of HO-1 can prevent ethanol induced cellular damage. Ethanol 172-179 heme oxygenase 1 Homo sapiens 155-159 15602829-4 2004 HO-1 activity was indicated by bilirubin and Fe2+ formation. Bilirubin 31-40 heme oxygenase 1 Homo sapiens 0-4 15602829-4 2004 HO-1 activity was indicated by bilirubin and Fe2+ formation. ammonium ferrous sulfate 45-49 heme oxygenase 1 Homo sapiens 0-4 15602829-6 2004 RESULTS: We first demonstrated a dose-dependent response between ethanol exposure and HO-1 mRNA and protein expression in human hepatocytes. Ethanol 65-72 heme oxygenase 1 Homo sapiens 86-90 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. Ethanol 87-94 heme oxygenase 1 Homo sapiens 49-53 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. Ethanol 121-128 heme oxygenase 1 Homo sapiens 49-53 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. Ethanol 121-128 heme oxygenase 1 Homo sapiens 49-53 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. Ethanol 121-128 heme oxygenase 1 Homo sapiens 49-53 15602829-7 2004 We further observed a time-dependent increase of HO-1 mRNA expression using 100 mmol/L ethanol starting 30 minutes after ethanol exposure, reaching its maximum between 3 h and 9 h. Being similar increased protein expression started to what had been demonstrated with the mRNA level, at 6 h after ethanol exposure, and kept continuous elevated over 18 h. In addition, we found that ethanol exposure to hepatocytes markedly increased HO-1 enzyme activity in a time-dependent manner measured as bilirubin and Fe2+ formation in human hepatocytes. Bilirubin 492-501 heme oxygenase 1 Homo sapiens 49-53 15518801-1 2004 To study the effect of treatment with cobalt-protoporphyrin (CoPP) for the induction of the heme oxygenase-1 (HO-1) enzyme on islet engraftment donor mice received either a single intraperitoneal injection of CoPP (20 mg/kg body weight) 1 day prior to islet isolation or this injection plus a 9 day posttransplantation course of Copp. cobaltiprotoporphyrin 38-59 heme oxygenase 1 Homo sapiens 92-108 15240748-7 2004 Also, inhibition of HO-1 activity by the specific inhibitor zinc-II-protoporphyrin-IX had no effect on the anti-inflammatory function of IL-10. zinc-ii-protoporphyrin-ix 60-85 heme oxygenase 1 Homo sapiens 20-24 15339982-8 2004 Heme catabolism is regulated by (inducible) heme oxygenase-1 (HO-1). Heme 0-4 heme oxygenase 1 Homo sapiens 44-60 15339982-8 2004 Heme catabolism is regulated by (inducible) heme oxygenase-1 (HO-1). Heme 0-4 heme oxygenase 1 Homo sapiens 62-66 15339982-10 2004 Stimulation of cells for 6 h with CdCl(2), which markedly increased HO-1 expression before the addition of VT, blunted subsequent hemin injury. cdcl 34-38 heme oxygenase 1 Homo sapiens 68-72 15339982-11 2004 In conclusion, VT augments hemin-induced toxicity in renal tubular epithelial cells that can be reversed by prior induction of HO-1. Hemin 27-32 heme oxygenase 1 Homo sapiens 127-131 15322212-5 2004 We demonstrate that aromatic and polar DEP fractions, which are enriched in polycyclic aromatic hydrocarbons and quinones, respectively, induce the expression of HO-1, GST, and other phase II enzymes in macrophages and epithelial cells. Polycyclic Aromatic Hydrocarbons 76-108 heme oxygenase 1 Homo sapiens 162-166 15322212-5 2004 We demonstrate that aromatic and polar DEP fractions, which are enriched in polycyclic aromatic hydrocarbons and quinones, respectively, induce the expression of HO-1, GST, and other phase II enzymes in macrophages and epithelial cells. Quinones 113-121 heme oxygenase 1 Homo sapiens 162-166 15518801-1 2004 To study the effect of treatment with cobalt-protoporphyrin (CoPP) for the induction of the heme oxygenase-1 (HO-1) enzyme on islet engraftment donor mice received either a single intraperitoneal injection of CoPP (20 mg/kg body weight) 1 day prior to islet isolation or this injection plus a 9 day posttransplantation course of Copp. cobaltiprotoporphyrin 38-59 heme oxygenase 1 Homo sapiens 110-114 15518801-1 2004 To study the effect of treatment with cobalt-protoporphyrin (CoPP) for the induction of the heme oxygenase-1 (HO-1) enzyme on islet engraftment donor mice received either a single intraperitoneal injection of CoPP (20 mg/kg body weight) 1 day prior to islet isolation or this injection plus a 9 day posttransplantation course of Copp. cobaltiprotoporphyrin 61-65 heme oxygenase 1 Homo sapiens 92-108 15518801-1 2004 To study the effect of treatment with cobalt-protoporphyrin (CoPP) for the induction of the heme oxygenase-1 (HO-1) enzyme on islet engraftment donor mice received either a single intraperitoneal injection of CoPP (20 mg/kg body weight) 1 day prior to islet isolation or this injection plus a 9 day posttransplantation course of Copp. cobaltiprotoporphyrin 61-65 heme oxygenase 1 Homo sapiens 110-114 15271722-2 2004 HO-1 catalyzes the conversion of the heme moiety of hemeproteins, such as hemoglobin, myoglobin, and cytochrome P450, to biliverdin, liberating carbon monoxide (CO) in the process. Heme 37-41 heme oxygenase 1 Homo sapiens 0-4 15311945-3 2004 We have observed that pretreatment of cells with PTIO boosted expression of IL-8 and heme oxygenase 1 (HOX) genes to a high level in cells treated with the NO donor DPTA-NO. dpta-no 165-172 heme oxygenase 1 Homo sapiens 85-101 15311945-3 2004 We have observed that pretreatment of cells with PTIO boosted expression of IL-8 and heme oxygenase 1 (HOX) genes to a high level in cells treated with the NO donor DPTA-NO. dpta-no 165-172 heme oxygenase 1 Homo sapiens 103-106 15311945-5 2004 The effect of PTIO was abrogated by reduced glutathione, suggesting that upregulation of the IL-8 and HOX genes is dependent on RNI-mediated S-nitrosation of specific regulator(s). Glutathione 44-55 heme oxygenase 1 Homo sapiens 102-105 15311945-6 2004 The concentration of PTIO required to enhance mRNA level was different for IL-8 and HOX genes. 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide 21-25 heme oxygenase 1 Homo sapiens 84-87 15258907-0 2004 Heme oxygenase-1 protects against apoptosis induced by tumor necrosis factor-alpha and cycloheximide in papillary thyroid carcinoma cells. Cycloheximide 87-100 heme oxygenase 1 Homo sapiens 0-16 15258907-3 2004 The results show that HO-1 is significantly induced by hemin and cadmium. Hemin 55-60 heme oxygenase 1 Homo sapiens 22-26 15258907-3 2004 The results show that HO-1 is significantly induced by hemin and cadmium. Cadmium 65-72 heme oxygenase 1 Homo sapiens 22-26 15258907-4 2004 In addition to inducing HO-1, hemin and cadmium also cause a rise in the levels of p21, a cyclin-dependent kinase inhibitor. Hemin 30-35 heme oxygenase 1 Homo sapiens 24-28 15258907-4 2004 In addition to inducing HO-1, hemin and cadmium also cause a rise in the levels of p21, a cyclin-dependent kinase inhibitor. Cadmium 40-47 heme oxygenase 1 Homo sapiens 24-28 15612472-0 2004 [Effect of acute ethanol exposure on HO-1 enzyme activity in human primary hepatocytes]. Ethanol 17-24 heme oxygenase 1 Homo sapiens 37-41 15612472-1 2004 OBJECTIVE: To study the effect of ethanol exposure on the expression of HO-1 enzymes in human primary culture hepatocytes. Ethanol 34-41 heme oxygenase 1 Homo sapiens 72-76 15612472-6 2004 Moreover, we also observed that ethanol exposure could affect HO-1 enzyme activity and HO-1 protein level, at the beginning, it went up significantly then went down. Ethanol 32-39 heme oxygenase 1 Homo sapiens 62-91 15612472-7 2004 CONCLUSION: The increase of HO-1 enzyme activity and protein level after ethanol exposure may be relative to prevent against ethanol-induced oxidative injury in human primary hepatocytes. Ethanol 73-80 heme oxygenase 1 Homo sapiens 28-32 15612472-7 2004 CONCLUSION: The increase of HO-1 enzyme activity and protein level after ethanol exposure may be relative to prevent against ethanol-induced oxidative injury in human primary hepatocytes. Ethanol 125-132 heme oxygenase 1 Homo sapiens 28-32 15271722-2 2004 HO-1 catalyzes the conversion of the heme moiety of hemeproteins, such as hemoglobin, myoglobin, and cytochrome P450, to biliverdin, liberating carbon monoxide (CO) in the process. Biliverdine 121-131 heme oxygenase 1 Homo sapiens 0-4 15271722-2 2004 HO-1 catalyzes the conversion of the heme moiety of hemeproteins, such as hemoglobin, myoglobin, and cytochrome P450, to biliverdin, liberating carbon monoxide (CO) in the process. Carbon Monoxide 144-159 heme oxygenase 1 Homo sapiens 0-4 15271722-2 2004 HO-1 catalyzes the conversion of the heme moiety of hemeproteins, such as hemoglobin, myoglobin, and cytochrome P450, to biliverdin, liberating carbon monoxide (CO) in the process. Carbon Monoxide 161-163 heme oxygenase 1 Homo sapiens 0-4 15271722-7 2004 These results suggest that oxidative stress caused by anesthesia and/or surgery may induce HO-1, which catalyzes heme to produce CO, leading to increased exhaled CO concentration. Heme 113-117 heme oxygenase 1 Homo sapiens 91-95 15271722-7 2004 These results suggest that oxidative stress caused by anesthesia and/or surgery may induce HO-1, which catalyzes heme to produce CO, leading to increased exhaled CO concentration. Carbon Monoxide 129-131 heme oxygenase 1 Homo sapiens 91-95 15231239-4 2004 These findings indicate aberrations in iron homeostasis that, we suspect, arise primarily from heme, since heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in AD, and mitochondria, since mitochondria turnover, mitochondrial DNA, and cytochrome C oxidative activity are all increased in AD. Biliverdine 185-195 heme oxygenase 1 Homo sapiens 107-123 15313669-4 2004 The recombinant plasmid pcDNA3-HO-1 was transfected into endothelial cells, and recombinant HO-1 was expressed in the endothelial cells under G418 selection. antibiotic G 418 142-146 heme oxygenase 1 Homo sapiens 92-96 15231239-4 2004 These findings indicate aberrations in iron homeostasis that, we suspect, arise primarily from heme, since heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in AD, and mitochondria, since mitochondria turnover, mitochondrial DNA, and cytochrome C oxidative activity are all increased in AD. Iron 39-43 heme oxygenase 1 Homo sapiens 107-123 15276427-11 2004 mRNA levels of heme oxygenase-1 (HO-1) were decreased by NAC in DMA(V)-exposed, but MMA(V)-exposed cells. N-myristoyl-alaninol 64-67 heme oxygenase 1 Homo sapiens 15-31 15298625-7 2004 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by trinitrobenzene sulphonic acid or dextran sulphate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the intestinal tract. trinitrobenzene sulphonic acid 133-163 heme oxygenase 1 Homo sapiens 45-49 15298625-7 2004 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by trinitrobenzene sulphonic acid or dextran sulphate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the intestinal tract. trinitrobenzene sulphonic acid 133-163 heme oxygenase 1 Homo sapiens 222-226 15231239-4 2004 These findings indicate aberrations in iron homeostasis that, we suspect, arise primarily from heme, since heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in AD, and mitochondria, since mitochondria turnover, mitochondrial DNA, and cytochrome C oxidative activity are all increased in AD. Heme 95-99 heme oxygenase 1 Homo sapiens 107-123 15264013-2 2004 The present study was carried out to examine the effect of dopamine on the expression of heme oxygenase-1 and -2 (HO-1 and HO-2) in human neuroblastomas (SK-N-SH cell line) and the effects of selegiline and antioxidants on this expression. Dopamine 59-67 heme oxygenase 1 Homo sapiens 89-112 15231239-4 2004 These findings indicate aberrations in iron homeostasis that, we suspect, arise primarily from heme, since heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in AD, and mitochondria, since mitochondria turnover, mitochondrial DNA, and cytochrome C oxidative activity are all increased in AD. Iron 176-180 heme oxygenase 1 Homo sapiens 107-123 15356994-3 2004 Curcumin possesses anti-inflammatory activity and is a potent inhibitor of reactive oxygen-generating enzymes such as lipoxygenase/cyclooxygenase, xanthine dehydrogenase/oxidase and inducible nitric oxide synthase; and an effective inducer of heme oxygenase-1. Curcumin 0-8 heme oxygenase 1 Homo sapiens 243-259 15356994-3 2004 Curcumin possesses anti-inflammatory activity and is a potent inhibitor of reactive oxygen-generating enzymes such as lipoxygenase/cyclooxygenase, xanthine dehydrogenase/oxidase and inducible nitric oxide synthase; and an effective inducer of heme oxygenase-1. reactive oxygen 75-90 heme oxygenase 1 Homo sapiens 243-259 15298625-7 2004 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by trinitrobenzene sulphonic acid or dextran sulphate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the intestinal tract. dextran sulphate sodium 167-190 heme oxygenase 1 Homo sapiens 45-49 15298625-7 2004 Recent studies suggest that the induction of HO-1 expression plays a critical protective role in intestinal damage models induced by trinitrobenzene sulphonic acid or dextran sulphate sodium, indicating that activation of HO-1 may act as an endogenous defensive mechanism to reduce inflammation and tissue injury in the intestinal tract. dextran sulphate sodium 167-190 heme oxygenase 1 Homo sapiens 222-226 15264013-2 2004 The present study was carried out to examine the effect of dopamine on the expression of heme oxygenase-1 and -2 (HO-1 and HO-2) in human neuroblastomas (SK-N-SH cell line) and the effects of selegiline and antioxidants on this expression. Dopamine 59-67 heme oxygenase 1 Homo sapiens 114-127 15264013-5 2004 The levels of HO-1 mRNA increased after dopamine treatment, in a dose-dependent manner, in all cell lines studied, whereas levels of the two HO-2 transcripts did not. Dopamine 40-48 heme oxygenase 1 Homo sapiens 14-18 15264013-7 2004 HO-1 protein was undetectable in untreated SK-N-SH cells and increased after treatment with dopamine. Dopamine 92-100 heme oxygenase 1 Homo sapiens 0-4 15264013-10 2004 Selegiline (10 microM) produced significant increase (P < 0.01) in the induction of HO-1 by dopamine (more than six times the control values). Selegiline 0-10 heme oxygenase 1 Homo sapiens 87-91 15264013-10 2004 Selegiline (10 microM) produced significant increase (P < 0.01) in the induction of HO-1 by dopamine (more than six times the control values). Dopamine 95-103 heme oxygenase 1 Homo sapiens 87-91 15264013-11 2004 The increased expression of HO-1 following dopamine treatment indicates that dopamine produces oxidative stress in this cell line. Dopamine 43-51 heme oxygenase 1 Homo sapiens 28-32 15264013-11 2004 The increased expression of HO-1 following dopamine treatment indicates that dopamine produces oxidative stress in this cell line. Dopamine 77-85 heme oxygenase 1 Homo sapiens 28-32 15233805-3 2004 HO-1 catalyzes heme breakdown to release iron, carbon monoxide, and biliverdin, which is reduced to bilirubin, a potent radical scavenger. Iron 41-45 heme oxygenase 1 Homo sapiens 0-4 15233805-3 2004 HO-1 catalyzes heme breakdown to release iron, carbon monoxide, and biliverdin, which is reduced to bilirubin, a potent radical scavenger. Carbon Monoxide 47-62 heme oxygenase 1 Homo sapiens 0-4 15233805-3 2004 HO-1 catalyzes heme breakdown to release iron, carbon monoxide, and biliverdin, which is reduced to bilirubin, a potent radical scavenger. Biliverdine 68-78 heme oxygenase 1 Homo sapiens 0-4 15233805-3 2004 HO-1 catalyzes heme breakdown to release iron, carbon monoxide, and biliverdin, which is reduced to bilirubin, a potent radical scavenger. Bilirubin 100-109 heme oxygenase 1 Homo sapiens 0-4 15213272-4 2004 SSAT overexpression increased the expression of heme oxygenase-1 (HO-1) by 350% 24 h after addition of tetracycline, indicating the induction of oxidative stress. Tetracycline 103-115 heme oxygenase 1 Homo sapiens 48-64 15213272-4 2004 SSAT overexpression increased the expression of heme oxygenase-1 (HO-1) by 350% 24 h after addition of tetracycline, indicating the induction of oxidative stress. Tetracycline 103-115 heme oxygenase 1 Homo sapiens 66-70 15213272-5 2004 The presence of catalase significantly prevented the upregulation of HO-1 in SSAT overexpressing cells, indicating that generation of H2O2 is partially responsible for the induction of oxidative stress. Hydrogen Peroxide 134-138 heme oxygenase 1 Homo sapiens 69-73 15219989-0 2004 Hydroxylamine and hydrazine bind directly to the heme iron of the heme-heme oxygenase-1 complex. Hydroxylamine 0-13 heme oxygenase 1 Homo sapiens 71-87 15219989-0 2004 Hydroxylamine and hydrazine bind directly to the heme iron of the heme-heme oxygenase-1 complex. hydrazine 18-27 heme oxygenase 1 Homo sapiens 71-87 15551655-11 2004 Dopamine is capable of stimulating the induction of protective enzymes like heme oxygenase-1 (HO-1) rendering the organ more resistant to the insult of ischemia/reperfusion and inflammation. Dopamine 0-8 heme oxygenase 1 Homo sapiens 76-92 15219989-0 2004 Hydroxylamine and hydrazine bind directly to the heme iron of the heme-heme oxygenase-1 complex. Heme 49-53 heme oxygenase 1 Homo sapiens 71-87 15219989-0 2004 Hydroxylamine and hydrazine bind directly to the heme iron of the heme-heme oxygenase-1 complex. Iron 54-58 heme oxygenase 1 Homo sapiens 71-87 15213303-0 2004 Roles of heme oxygenase-1 in the antiproliferative and antiapoptotic effects of nitric oxide on Jurkat T cells. Nitric Oxide 80-92 heme oxygenase 1 Homo sapiens 9-25 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Heme 40-44 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Heme 40-44 heme oxygenase 1 Homo sapiens 18-22 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Biliverdine 50-60 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Biliverdine 50-60 heme oxygenase 1 Homo sapiens 18-22 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Iron 67-71 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Iron 67-71 heme oxygenase 1 Homo sapiens 18-22 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. ammonium ferrous sulfate 73-79 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. ammonium ferrous sulfate 73-79 heme oxygenase 1 Homo sapiens 18-22 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Carbon Monoxide 86-101 heme oxygenase 1 Homo sapiens 18-22 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Carbon Monoxide 103-105 heme oxygenase 1 Homo sapiens 0-16 15213303-2 2004 Heme oxygenase-1 (HO-1), which degrades heme into biliverdin, free iron (Fe(2+)), and carbon monoxide (CO), has also been known to have antiproliferative and antiapoptotic effects. Carbon Monoxide 103-105 heme oxygenase 1 Homo sapiens 18-22 15213303-5 2004 Using human Jurkat T cells, we found that the NO donor sodium nitroprusside (SNP) induced HO-1 expression and that preincubation with SNP suppressed T cell proliferation induced by concanavalin A and apoptosis triggered by anti-Fas antibody. Nitroprusside 55-75 heme oxygenase 1 Homo sapiens 90-94 15213303-6 2004 Suppressions of T cell proliferation and apoptosis comparable with SNP were also observed when the T cells were preincubated with the HO-1 inducer cobalt protoporphyrin. cobaltiprotoporphyrin 147-168 heme oxygenase 1 Homo sapiens 134-138 15213303-7 2004 A phosphorothioate-linked HO-1 antisense oligonucleotide blocked HO-1 expression, and subsequently abrogated the antiproliferative and antiapoptotic effects of SNP. Parathion 2-18 heme oxygenase 1 Homo sapiens 26-30 15213303-7 2004 A phosphorothioate-linked HO-1 antisense oligonucleotide blocked HO-1 expression, and subsequently abrogated the antiproliferative and antiapoptotic effects of SNP. Parathion 2-18 heme oxygenase 1 Homo sapiens 65-69 15213303-7 2004 A phosphorothioate-linked HO-1 antisense oligonucleotide blocked HO-1 expression, and subsequently abrogated the antiproliferative and antiapoptotic effects of SNP. Oligonucleotides 41-56 heme oxygenase 1 Homo sapiens 26-30 15213303-7 2004 A phosphorothioate-linked HO-1 antisense oligonucleotide blocked HO-1 expression, and subsequently abrogated the antiproliferative and antiapoptotic effects of SNP. Oligonucleotides 41-56 heme oxygenase 1 Homo sapiens 65-69 15188507-7 2004 The specific HO-1 inhibitor- ZnPPIX could worsen LPS-induced injuries and weaken the protective effect of CCK-8. zinc protoporphyrin 29-35 heme oxygenase 1 Homo sapiens 13-17 15207648-4 2004 Immunoblot experiments showed that Pb2+ exposure (100 nM to 10 microM, 1-14 days) induces HO-1 synthesis in astrocytes, but not in neurons; this is probably the 32-kDa protein. Lead 35-39 heme oxygenase 1 Homo sapiens 90-94 15207648-7 2004 We found that HO-1 induction in astrocytes is increased by combined exposure to Pb2+ and many other stresses, including heat, nitric oxide, H2O2, and superoxide. Nitric Oxide 126-138 heme oxygenase 1 Homo sapiens 14-18 15207648-7 2004 We found that HO-1 induction in astrocytes is increased by combined exposure to Pb2+ and many other stresses, including heat, nitric oxide, H2O2, and superoxide. Hydrogen Peroxide 140-144 heme oxygenase 1 Homo sapiens 14-18 15207648-7 2004 We found that HO-1 induction in astrocytes is increased by combined exposure to Pb2+ and many other stresses, including heat, nitric oxide, H2O2, and superoxide. Superoxides 150-160 heme oxygenase 1 Homo sapiens 14-18 15207648-10 2004 Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). Lead 21-25 heme oxygenase 1 Homo sapiens 13-17 15207648-10 2004 Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). Hydroxyl Radical 44-60 heme oxygenase 1 Homo sapiens 13-17 15207648-10 2004 Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). 1,3-dimethylthiourea 72-88 heme oxygenase 1 Homo sapiens 13-17 15207648-10 2004 Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). 1,3-dimethylthiourea 90-94 heme oxygenase 1 Homo sapiens 13-17 15207648-10 2004 Induction of HO-1 by Pb2+ is reduced by the hydroxyl radical scavengers dimethylthiourea (DMTU) and mannitol, but not by inhibitors of calmodulin, calmodulin-dependent protein kinases, protein kinase C, or extracellular signal-regulated kinases (ERK). Mannitol 100-108 heme oxygenase 1 Homo sapiens 13-17 15188507-7 2004 The specific HO-1 inhibitor- ZnPPIX could worsen LPS-induced injuries and weaken the protective effect of CCK-8. Sincalide 106-111 heme oxygenase 1 Homo sapiens 13-17 15188507-9 2004 CONCLUSION: HO-1 may be a key factor in CCK-8 attenuated injuries of PASMCs induced by LPS, and HO-1 expression may be related to the activation of JNK and activator protein (AP-1). pasmcs 69-75 heme oxygenase 1 Homo sapiens 12-16 15161651-6 2004 A rapid increase in the expression of genes involved in reactive oxygen species metabolism and cell stress, eg, heme oxygenase-1, thioredoxin reductase, cytochrome b5 reductase, Gadd 153, amino acid transporter E16, and HSP70 was found after HOCl-LDL treatment of HK-2 cells. Reactive Oxygen Species 56-79 heme oxygenase 1 Homo sapiens 112-128 15024026-0 2004 15-deoxy-delta 12,14-prostaglandin J2 induces heme oxygenase-1 gene expression in a reactive oxygen species-dependent manner in human lymphocytes. 15-deoxy-delta(12,14)-prostaglandin J2 0-37 heme oxygenase 1 Homo sapiens 46-62 15180563-3 2004 Exposure of mammalian cells to oxidative stimuli induces heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, as well as a 33-kDa heat shock protein. Heme 57-61 heme oxygenase 1 Homo sapiens 75-79 15158123-3 2004 In this study, we found that a 30-min incubation in the absence of glucose resulted in a rapid increased expression of COX-2 and HO-1 in cardiac fibroblasts as examined by real-time quantitative polymerase chain reaction (PCR) and western blot analysis. Glucose 67-74 heme oxygenase 1 Homo sapiens 129-133 15158123-7 2004 In addition, the induction of COX-2 and HO-1 expression by TGD was prevented by pretreatment with NAC or SB203580, a p38 MAPK inhibitor. Acetylcysteine 98-101 heme oxygenase 1 Homo sapiens 40-44 15158123-7 2004 In addition, the induction of COX-2 and HO-1 expression by TGD was prevented by pretreatment with NAC or SB203580, a p38 MAPK inhibitor. SB 203580 105-113 heme oxygenase 1 Homo sapiens 40-44 15158123-8 2004 Surprisingly, pretreatment with chelerythrine, an inhibitor of PKC, strongly augmented the HO-1 mRNA expression but blocked the COX-2 mRNA induction by TGD. chelerythrine 32-45 heme oxygenase 1 Homo sapiens 91-95 15158123-9 2004 These results demonstrate that briefly removing glucose from cultured cardiac fibroblasts induces COX-2 and HO-1 expression via generation of ROS and p38 MAPK phosphorylation, while the translocation of PKC- to the membrane fraction may participate in the induction of COX-2 but not in the HO-1 expression. Glucose 48-55 heme oxygenase 1 Homo sapiens 108-112 15158123-9 2004 These results demonstrate that briefly removing glucose from cultured cardiac fibroblasts induces COX-2 and HO-1 expression via generation of ROS and p38 MAPK phosphorylation, while the translocation of PKC- to the membrane fraction may participate in the induction of COX-2 but not in the HO-1 expression. Glucose 48-55 heme oxygenase 1 Homo sapiens 290-294 15158123-9 2004 These results demonstrate that briefly removing glucose from cultured cardiac fibroblasts induces COX-2 and HO-1 expression via generation of ROS and p38 MAPK phosphorylation, while the translocation of PKC- to the membrane fraction may participate in the induction of COX-2 but not in the HO-1 expression. Reactive Oxygen Species 142-145 heme oxygenase 1 Homo sapiens 108-112 15158359-9 2004 These results showing a CO-mediated decrease in 59Fe uptake from 59Fe-Tf using exogenous CO were in agreement with studies implementing cells transfected with HO1. Iron-59 48-52 heme oxygenase 1 Homo sapiens 159-162 15024026-0 2004 15-deoxy-delta 12,14-prostaglandin J2 induces heme oxygenase-1 gene expression in a reactive oxygen species-dependent manner in human lymphocytes. Reactive Oxygen Species 84-107 heme oxygenase 1 Homo sapiens 46-62 15024026-6 2004 Inhibition of intracellular ROS production by N-acetylcysteine, TEMPO, Me(2)SO, 1,10-phenanthroline, or allopurinol resulted in a decreased 15dPGJ(2)-dependent HO-1 expression in the cells. Reactive Oxygen Species 28-31 heme oxygenase 1 Homo sapiens 160-164 15024026-6 2004 Inhibition of intracellular ROS production by N-acetylcysteine, TEMPO, Me(2)SO, 1,10-phenanthroline, or allopurinol resulted in a decreased 15dPGJ(2)-dependent HO-1 expression in the cells. Acetylcysteine 46-62 heme oxygenase 1 Homo sapiens 160-164 15024026-6 2004 Inhibition of intracellular ROS production by N-acetylcysteine, TEMPO, Me(2)SO, 1,10-phenanthroline, or allopurinol resulted in a decreased 15dPGJ(2)-dependent HO-1 expression in the cells. 1,10-phenanthroline 80-99 heme oxygenase 1 Homo sapiens 160-164 15024026-6 2004 Inhibition of intracellular ROS production by N-acetylcysteine, TEMPO, Me(2)SO, 1,10-phenanthroline, or allopurinol resulted in a decreased 15dPGJ(2)-dependent HO-1 expression in the cells. Allopurinol 104-115 heme oxygenase 1 Homo sapiens 160-164 15024026-7 2004 Furthermore, buthionine sulfoximine, an inhibitor of reduced glutathione synthesis, or Fe(2+)/Cu(2+) ions enhanced the positive effect of 15dPGJ(2) on HO-1 expression. Buthionine Sulfoximine 13-35 heme oxygenase 1 Homo sapiens 151-155 15024026-7 2004 Furthermore, buthionine sulfoximine, an inhibitor of reduced glutathione synthesis, or Fe(2+)/Cu(2+) ions enhanced the positive effect of 15dPGJ(2) on HO-1 expression. Glutathione 61-72 heme oxygenase 1 Homo sapiens 151-155 15024026-7 2004 Furthermore, buthionine sulfoximine, an inhibitor of reduced glutathione synthesis, or Fe(2+)/Cu(2+) ions enhanced the positive effect of 15dPGJ(2) on HO-1 expression. ammonium ferrous sulfate 87-93 heme oxygenase 1 Homo sapiens 151-155 15024026-7 2004 Furthermore, buthionine sulfoximine, an inhibitor of reduced glutathione synthesis, or Fe(2+)/Cu(2+) ions enhanced the positive effect of 15dPGJ(2) on HO-1 expression. cupric ion 94-100 heme oxygenase 1 Homo sapiens 151-155 15084179-2 2004 The induction of HO-1 expression is modulated by a (GT)(n) dinucleotide polymorphism in the promoter of the gene, of which increased activity is associated with short (S) (<or=27) repeats. (n) dinucleotide 55-71 heme oxygenase 1 Homo sapiens 17-21 14988408-14 2004 hBVR modulation of ATF-2 and HO-1 expression suggests it has a potential role in regulation of AP-1 and cAMP-regulated genes and a role in cell signaling. Cyclic AMP 104-108 heme oxygenase 1 Homo sapiens 29-33 14731112-9 2004 The half-life of HO-1 protein was prolonged by MG132, indicating that the upregulation of HO-1 by proteasome inhibitor is partially attributable to the inhibition of protein degradation. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 47-52 heme oxygenase 1 Homo sapiens 90-94 14731112-11 2004 We found HO-1 upregulation by MG132 and pyrrolidine dithiocarbamate is unrelated to their inhibition of NF-kappaB, since leptomycin B, another NF-kappaB inhibitor, did not elicit similar induction of HO-1. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 30-35 heme oxygenase 1 Homo sapiens 9-13 14731112-3 2004 In the present study, we found a novel action of proteasome inhibitors MG132 and MG262 on HO-1 induction, and characterized the underlying mechanisms. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 71-76 heme oxygenase 1 Homo sapiens 90-94 14731112-11 2004 We found HO-1 upregulation by MG132 and pyrrolidine dithiocarbamate is unrelated to their inhibition of NF-kappaB, since leptomycin B, another NF-kappaB inhibitor, did not elicit similar induction of HO-1. pyrrolidine dithiocarbamic acid 40-67 heme oxygenase 1 Homo sapiens 9-13 14731112-3 2004 In the present study, we found a novel action of proteasome inhibitors MG132 and MG262 on HO-1 induction, and characterized the underlying mechanisms. MG 262 81-86 heme oxygenase 1 Homo sapiens 90-94 14731112-4 2004 MG132 (> or =0.1 microM) treatment resulted in a marked time- and concentration-dependent induction of the steady-state level of HO-1 mRNA in RAW264.7 macrophages, followed by a corresponding increase in HO-1 protein. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 heme oxygenase 1 Homo sapiens 132-136 14731112-4 2004 MG132 (> or =0.1 microM) treatment resulted in a marked time- and concentration-dependent induction of the steady-state level of HO-1 mRNA in RAW264.7 macrophages, followed by a corresponding increase in HO-1 protein. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 0-5 heme oxygenase 1 Homo sapiens 207-211 14731112-5 2004 Actinomycin D and cycloheximide inhibited MG132-responsive HO-1 protein expression, indicating a requirement for transcription and de novo protein synthesis. Dactinomycin 0-13 heme oxygenase 1 Homo sapiens 59-63 14731112-5 2004 Actinomycin D and cycloheximide inhibited MG132-responsive HO-1 protein expression, indicating a requirement for transcription and de novo protein synthesis. Cycloheximide 18-31 heme oxygenase 1 Homo sapiens 59-63 14731112-5 2004 Actinomycin D and cycloheximide inhibited MG132-responsive HO-1 protein expression, indicating a requirement for transcription and de novo protein synthesis. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 42-47 heme oxygenase 1 Homo sapiens 59-63 14731112-6 2004 The involvement of signal pathways in MG132-induced HO-1 gene expression was examined using chemical inhibitors. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 38-43 heme oxygenase 1 Homo sapiens 52-56 14731112-7 2004 Antioxidant N -acetylcysteine and SB203580, an antioxidant and inhibitor of p38 MAPK (mitogen-activated protein kinase), abolished MG132-inducible HO-1 expression. Acetylcysteine 12-29 heme oxygenase 1 Homo sapiens 147-151 14731112-7 2004 Antioxidant N -acetylcysteine and SB203580, an antioxidant and inhibitor of p38 MAPK (mitogen-activated protein kinase), abolished MG132-inducible HO-1 expression. SB 203580 34-42 heme oxygenase 1 Homo sapiens 147-151 15190962-1 2004 OBJECTIVE: We aimed to quantify concentrations of inducible heme oxygenase (HO)-1 in the lungs of patients with acute respiratory distress syndrome (ARDS) and to investigate its role as a source of ferrous iron and as a signaling agent for iron regulation. Iron 206-210 heme oxygenase 1 Homo sapiens 60-81 14731112-7 2004 Antioxidant N -acetylcysteine and SB203580, an antioxidant and inhibitor of p38 MAPK (mitogen-activated protein kinase), abolished MG132-inducible HO-1 expression. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 131-136 heme oxygenase 1 Homo sapiens 147-151 14731112-9 2004 The half-life of HO-1 protein was prolonged by MG132, indicating that the upregulation of HO-1 by proteasome inhibitor is partially attributable to the inhibition of protein degradation. benzyloxycarbonylleucyl-leucyl-leucine aldehyde 47-52 heme oxygenase 1 Homo sapiens 17-21 15126353-2 2004 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the degradation of heme and has recently been implicated in the regulation of growth and survival of various neoplastic cells. Heme 89-93 heme oxygenase 1 Homo sapiens 0-16 15126353-2 2004 Heme oxygenase-1 (HO-1) is an inducible stress protein that catalyzes the degradation of heme and has recently been implicated in the regulation of growth and survival of various neoplastic cells. Heme 89-93 heme oxygenase 1 Homo sapiens 18-22 15126353-5 2004 Exposure of these cells to the BCR/ABL tyrosine kinase inhibitor STI571 resulted in decreased expression of HO-1 mRNA and protein. Imatinib Mesylate 65-71 heme oxygenase 1 Homo sapiens 108-112 15126353-8 2004 Hemin-induced expression of HO-1 was found to protect CML cells from STI571-induced cell death. Imatinib Mesylate 69-75 heme oxygenase 1 Homo sapiens 28-32 15126353-9 2004 In addition, inhibition of HO-1 by zinc-(II)-deuteroporphyrin-IX-2,4-bisethyleneglycol resulted in a substantial decrease of cell viability. zinc-(ii)-deuteroporphyrin-ix-2,4-bisethyleneglycol 35-86 heme oxygenase 1 Homo sapiens 27-31 14726403-8 2004 Increased HO-1 enzymatic activity in vitro enhanced proliferation of KSHV-infected DMVECs in the presence of free heme. dmvecs 83-89 heme oxygenase 1 Homo sapiens 10-14 14726403-8 2004 Increased HO-1 enzymatic activity in vitro enhanced proliferation of KSHV-infected DMVECs in the presence of free heme. Heme 114-118 heme oxygenase 1 Homo sapiens 10-14 14726403-9 2004 Treatment with the HO-1 inhibitor chromium mesoporphyrin IX abolished heme-induced proliferation. chromium mesoporphyrin ix 34-59 heme oxygenase 1 Homo sapiens 19-23 14726403-9 2004 Treatment with the HO-1 inhibitor chromium mesoporphyrin IX abolished heme-induced proliferation. Heme 70-74 heme oxygenase 1 Homo sapiens 19-23 15126353-10 2004 Furthermore, overexpression of HO-1 in the CML-derived cell line K562 was found to counteract STI571-induced apoptosis. Imatinib Mesylate 94-100 heme oxygenase 1 Homo sapiens 31-35 15190962-1 2004 OBJECTIVE: We aimed to quantify concentrations of inducible heme oxygenase (HO)-1 in the lungs of patients with acute respiratory distress syndrome (ARDS) and to investigate its role as a source of ferrous iron and as a signaling agent for iron regulation. Iron 240-244 heme oxygenase 1 Homo sapiens 60-81 15086901-7 2004 In TALH cells infected with Ad-NKCC2-HO-1, Ang II-stimulated prostaglandin E(2) (PGE(2)) levels were reduced by 40%. Dinoprostone 61-79 heme oxygenase 1 Homo sapiens 37-41 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 15-20 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 36-52 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 54-58 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 140-144 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 237-246 heme oxygenase 1 Homo sapiens 15-20 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 237-246 heme oxygenase 1 Homo sapiens 36-52 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 237-246 heme oxygenase 1 Homo sapiens 54-58 15341181-8 2004 In particular, HSP32, also known as heme oxygenase-1 (HO-1), has received considerable attention, as it has been recently demonstrated that HO-1 induction, by generating the vasoactive molecule carbon monoxide and the potent antioxidant bilirubin, could represent a protective system potentially active against brain oxidative injury. Bilirubin 237-246 heme oxygenase 1 Homo sapiens 140-144 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Heme 64-68 heme oxygenase 1 Homo sapiens 12-28 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Heme 64-68 heme oxygenase 1 Homo sapiens 30-34 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Bilirubin 72-81 heme oxygenase 1 Homo sapiens 12-28 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Bilirubin 72-81 heme oxygenase 1 Homo sapiens 30-34 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 12-28 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Carbon Monoxide 83-98 heme oxygenase 1 Homo sapiens 30-34 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Carbon Monoxide 100-102 heme oxygenase 1 Homo sapiens 12-28 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Carbon Monoxide 100-102 heme oxygenase 1 Homo sapiens 30-34 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Iron 114-118 heme oxygenase 1 Homo sapiens 12-28 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Iron 114-118 heme oxygenase 1 Homo sapiens 30-34 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Heme 159-163 heme oxygenase 1 Homo sapiens 12-28 15086901-7 2004 In TALH cells infected with Ad-NKCC2-HO-1, Ang II-stimulated prostaglandin E(2) (PGE(2)) levels were reduced by 40%. Prostaglandins E 81-84 heme oxygenase 1 Homo sapiens 37-41 15086901-1 2004 BACKGROUND: Heme oxygenase-1 (HO-1) catalyzes the conversion of heme to bilirubin, carbon monoxide (CO), and free iron, thus controlling the level of cellular heme. Heme 159-163 heme oxygenase 1 Homo sapiens 30-34 15086901-8 2004 Ang II caused a marked decrease in GSH levels and this decrease was greatly attenuated in TALH cells transduced with Ad-NKCC2-HO-1. Glutathione 35-38 heme oxygenase 1 Homo sapiens 126-130 15086901-3 2004 We hypothesize that selective upregulation of HO-1 in TALH by gene transfer attenuates oxidative stress caused by angiotensin II (Ang II). talh 54-58 heme oxygenase 1 Homo sapiens 46-50 14977878-1 2004 Biliverdin, a product of heme oxygenase-1 (HO-1) enzymatic action, is converted into bilirubin, which has been considered a waste product in the past. Biliverdine 0-10 heme oxygenase 1 Homo sapiens 25-41 15067050-0 2004 Carbon monoxide produced by heme oxygenase-1 suppresses T cell proliferation via inhibition of IL-2 production. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 28-44 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Heme 36-40 heme oxygenase 1 Homo sapiens 0-16 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Heme 36-40 heme oxygenase 1 Homo sapiens 18-22 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Carbon Monoxide 46-48 heme oxygenase 1 Homo sapiens 0-16 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Carbon Monoxide 46-48 heme oxygenase 1 Homo sapiens 18-22 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Biliverdine 50-60 heme oxygenase 1 Homo sapiens 0-16 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Biliverdine 50-60 heme oxygenase 1 Homo sapiens 18-22 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Iron 71-75 heme oxygenase 1 Homo sapiens 0-16 15067050-1 2004 Heme oxygenase-1 (HO-1) catabolizes heme into CO, biliverdin, and free iron and serves as a protective enzyme by virtue of its anti-inflammatory, antiapoptotic, and antiproliferative actions. Iron 71-75 heme oxygenase 1 Homo sapiens 18-22 15049686-0 2004 Crystal structure of human heme oxygenase-1 in a complex with biliverdin. Biliverdine 62-72 heme oxygenase 1 Homo sapiens 27-43 14977878-1 2004 Biliverdin, a product of heme oxygenase-1 (HO-1) enzymatic action, is converted into bilirubin, which has been considered a waste product in the past. Bilirubin 85-94 heme oxygenase 1 Homo sapiens 25-41 14977880-0 2004 Heme oxygenase-1-derived carbon monoxide protects hearts from transplant associated ischemia reperfusion injury. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Heme 33-37 heme oxygenase 1 Homo sapiens 0-16 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Heme 33-37 heme oxygenase 1 Homo sapiens 18-22 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Iron 43-47 heme oxygenase 1 Homo sapiens 0-16 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Heme 23-27 heme oxygenase 1 Homo sapiens 41-45 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Iron 43-47 heme oxygenase 1 Homo sapiens 18-22 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Carbon Monoxide 96-111 heme oxygenase 1 Homo sapiens 23-39 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Carbon Monoxide 96-111 heme oxygenase 1 Homo sapiens 41-45 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Carbon Monoxide 113-115 heme oxygenase 1 Homo sapiens 23-39 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Carbon Monoxide 113-115 heme oxygenase 1 Homo sapiens 41-45 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Biliverdine 118-129 heme oxygenase 1 Homo sapiens 23-39 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Biliverdine 118-129 heme oxygenase 1 Homo sapiens 41-45 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Iron 139-143 heme oxygenase 1 Homo sapiens 23-39 14973545-1 2004 Elevated expression of heme oxygenase-1 (HO-1), an intracellular enzyme that degrades heme into carbon monoxide (CO), biliverdine and free iron, has anti-inflammatory and antiapoptotic effects in diverse models. Iron 139-143 heme oxygenase 1 Homo sapiens 41-45 15019971-0 2004 Overexpression of heme oxygenase (HO)-1 renders Jurkat T cells resistant to fas-mediated apoptosis: involvement of iron released by HO-1. ammonium ferrous sulfate 76-79 heme oxygenase 1 Homo sapiens 18-39 15019971-0 2004 Overexpression of heme oxygenase (HO)-1 renders Jurkat T cells resistant to fas-mediated apoptosis: involvement of iron released by HO-1. Iron 115-119 heme oxygenase 1 Homo sapiens 18-39 15019971-0 2004 Overexpression of heme oxygenase (HO)-1 renders Jurkat T cells resistant to fas-mediated apoptosis: involvement of iron released by HO-1. Iron 115-119 heme oxygenase 1 Homo sapiens 132-136 15019971-4 2004 Surprisingly, however, HO-1-overexpressing Jurkat T cells showed strong resistance to Fas-mediated apoptosis. ammonium ferrous sulfate 86-89 heme oxygenase 1 Homo sapiens 23-27 15019971-5 2004 In contrast, abrogation of HO-1 expression by antisense oligomer against HO-1 gene from CoPP-treated cells or depletion of iron by desferrioxamine from HO-1-transfected cells abolished the resistance. cobaltiprotoporphyrin 88-92 heme oxygenase 1 Homo sapiens 27-31 15019971-5 2004 In contrast, abrogation of HO-1 expression by antisense oligomer against HO-1 gene from CoPP-treated cells or depletion of iron by desferrioxamine from HO-1-transfected cells abolished the resistance. cobaltiprotoporphyrin 88-92 heme oxygenase 1 Homo sapiens 73-77 15019971-5 2004 In contrast, abrogation of HO-1 expression by antisense oligomer against HO-1 gene from CoPP-treated cells or depletion of iron by desferrioxamine from HO-1-transfected cells abolished the resistance. cobaltiprotoporphyrin 88-92 heme oxygenase 1 Homo sapiens 73-77 15019971-8 2004 Primary CD4+ T cells induced by CoPP to express HO-1 also showed more resistance to Fas-mediated apoptosis than untreated cells. cobaltiprotoporphyrin 32-36 heme oxygenase 1 Homo sapiens 48-52 15019971-8 2004 Primary CD4+ T cells induced by CoPP to express HO-1 also showed more resistance to Fas-mediated apoptosis than untreated cells. ammonium ferrous sulfate 84-87 heme oxygenase 1 Homo sapiens 48-52 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Biliverdine 49-59 heme oxygenase 1 Homo sapiens 0-16 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Biliverdine 49-59 heme oxygenase 1 Homo sapiens 18-22 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 65-80 heme oxygenase 1 Homo sapiens 0-16 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 65-80 heme oxygenase 1 Homo sapiens 18-22 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 82-84 heme oxygenase 1 Homo sapiens 0-16 14977880-1 2004 Heme oxygenase-1 (HO-1) degrades heme into iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 82-84 heme oxygenase 1 Homo sapiens 18-22 14977880-4 2004 Induction of HO-1 expression by administration of cobalt protoporphyrin IX (CoPPIX) to the graft donor restored graft function. cobaltiprotoporphyrin 50-74 heme oxygenase 1 Homo sapiens 13-17 14977880-4 2004 Induction of HO-1 expression by administration of cobalt protoporphyrin IX (CoPPIX) to the graft donor restored graft function. cobaltiprotoporphyrin 76-82 heme oxygenase 1 Homo sapiens 13-17 14977880-5 2004 Inhibition of HO-1 enzymatic activity, by administration of zinc protoporphyrin (ZnPPIX) at the time of transplantation, reversed the protective effect of HO-1. zinc protoporphyrin 60-79 heme oxygenase 1 Homo sapiens 14-18 14977880-5 2004 Inhibition of HO-1 enzymatic activity, by administration of zinc protoporphyrin (ZnPPIX) at the time of transplantation, reversed the protective effect of HO-1. zinc protoporphyrin 60-79 heme oxygenase 1 Homo sapiens 155-159 14977880-5 2004 Inhibition of HO-1 enzymatic activity, by administration of zinc protoporphyrin (ZnPPIX) at the time of transplantation, reversed the protective effect of HO-1. zinc protoporphyrin 81-87 heme oxygenase 1 Homo sapiens 14-18 14977880-5 2004 Inhibition of HO-1 enzymatic activity, by administration of zinc protoporphyrin (ZnPPIX) at the time of transplantation, reversed the protective effect of HO-1. zinc protoporphyrin 81-87 heme oxygenase 1 Homo sapiens 155-159 15085064-7 2004 Manidipine increased monocyte HO-1 mRNA production (1.6 +/- 0.4 versus 1.2 +/- 0.4, P < 0.008), while nifedipine and captopril showed no effect (1.2 +/- 0.3 and 1.1 +/- 0.3, respectively). manidipine 0-10 heme oxygenase 1 Homo sapiens 30-34 15085064-0 2004 Effect of manidipine on gene expression and protein level of oxidative stress-related proteins: p22phox and HO-1: relevance for antihypertensive and anti-remodeling effects. manidipine 10-20 heme oxygenase 1 Homo sapiens 108-112 15085064-9 2004 In conclusion, manidipine decreases p22(phox) and increases HO-1 mRNA production and protein level. manidipine 15-25 heme oxygenase 1 Homo sapiens 60-64 15085064-10 2004 The manidipine-induced increase of HO-1 gene and protein expression seems to be a peculiar effect of this drug since it is not observed with captopril and nifedipine. manidipine 4-14 heme oxygenase 1 Homo sapiens 35-39 15085064-10 2004 The manidipine-induced increase of HO-1 gene and protein expression seems to be a peculiar effect of this drug since it is not observed with captopril and nifedipine. Captopril 141-150 heme oxygenase 1 Homo sapiens 35-39 15010533-7 2004 Inhibition of the Fenton reaction by the.OH scavenger DHR attenuated HIF-prolyl hydroxylase activity and interaction with von Hippel-Lindau protein, leading to enhanced HIF-1alpha levels, HIF-1alpha transactivation, and activated expression of the HIF-1 target genes plasminogen activator inhibitor 1 and heme oxygenase 1. dhr 54-57 heme oxygenase 1 Homo sapiens 305-321 14998722-0 2004 Thiol antioxidant and thiol-reducing agents attenuate 15-deoxy-delta 12,14-prostaglandin J2-induced heme oxygenase-1 expression. Sulfhydryl Compounds 0-5 heme oxygenase 1 Homo sapiens 100-116 14998722-0 2004 Thiol antioxidant and thiol-reducing agents attenuate 15-deoxy-delta 12,14-prostaglandin J2-induced heme oxygenase-1 expression. Sulfhydryl Compounds 22-27 heme oxygenase 1 Homo sapiens 100-116 14998722-0 2004 Thiol antioxidant and thiol-reducing agents attenuate 15-deoxy-delta 12,14-prostaglandin J2-induced heme oxygenase-1 expression. 15-deoxy-delta(12,14)-prostaglandin J2 54-91 heme oxygenase 1 Homo sapiens 100-116 14998722-2 2004 Herein we examined how various eicosanoids affect the induction of HO-1, and the possible mechanism underlying 15-deoxy-Delta(12,14)- prostaglandin J(2) (15d-PGJ(2))-induced HO-1 expression. Eicosanoids 31-42 heme oxygenase 1 Homo sapiens 67-71 14998722-2 2004 Herein we examined how various eicosanoids affect the induction of HO-1, and the possible mechanism underlying 15-deoxy-Delta(12,14)- prostaglandin J(2) (15d-PGJ(2))-induced HO-1 expression. 15-deoxy-delta 111-125 heme oxygenase 1 Homo sapiens 174-178 14998722-2 2004 Herein we examined how various eicosanoids affect the induction of HO-1, and the possible mechanism underlying 15-deoxy-Delta(12,14)- prostaglandin J(2) (15d-PGJ(2))-induced HO-1 expression. prostaglandin j 134-149 heme oxygenase 1 Homo sapiens 174-178 14998722-3 2004 PGH(2), PGD(2) and its metabolites of the PGJ(2) series, and PGA(1) markedly induced the protein expression of HO-1. GH2 protein, human 0-3 heme oxygenase 1 Homo sapiens 111-115 14998722-3 2004 PGH(2), PGD(2) and its metabolites of the PGJ(2) series, and PGA(1) markedly induced the protein expression of HO-1. Prostaglandins D 8-11 heme oxygenase 1 Homo sapiens 111-115 14998722-3 2004 PGH(2), PGD(2) and its metabolites of the PGJ(2) series, and PGA(1) markedly induced the protein expression of HO-1. 2-(ETHOXYMETHYL)-4-(4-FLUOROPHENYL)-3-[2-(2-HYDROXYPHENOXY)PYRIMIDIN-4-YL]ISOXAZOL-5(2H)-ONE 42-45 heme oxygenase 1 Homo sapiens 111-115 14998722-3 2004 PGH(2), PGD(2) and its metabolites of the PGJ(2) series, and PGA(1) markedly induced the protein expression of HO-1. Prostaglandins A 61-64 heme oxygenase 1 Homo sapiens 111-115 14998722-8 2004 15d-PGJ(2) significantly decreased the intracellular level of reduced glutathione; and the thiol antioxidant, N-acetyl-L-cysteine (NAC), and the thiol-reducing agent, dithiothreitol (DTT), inhibited the induction of HO-1 by 15d-PGJ(2). 15d-pgj 0-7 heme oxygenase 1 Homo sapiens 216-220 14998722-9 2004 Finally, NAC and DTT exhibited significant inhibition of HO-1 mRNA and HO-1 promoter reporter activity induced by 15d-PGJ(2). Dithiothreitol 17-20 heme oxygenase 1 Homo sapiens 57-61 14998722-9 2004 Finally, NAC and DTT exhibited significant inhibition of HO-1 mRNA and HO-1 promoter reporter activity induced by 15d-PGJ(2). Dithiothreitol 17-20 heme oxygenase 1 Homo sapiens 71-75 14998722-9 2004 Finally, NAC and DTT exhibited significant inhibition of HO-1 mRNA and HO-1 promoter reporter activity induced by 15d-PGJ(2). 15d-pgj 114-121 heme oxygenase 1 Homo sapiens 57-61 14998722-9 2004 Finally, NAC and DTT exhibited significant inhibition of HO-1 mRNA and HO-1 promoter reporter activity induced by 15d-PGJ(2). 15d-pgj 114-121 heme oxygenase 1 Homo sapiens 71-75 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. Sulfhydryl Compounds 27-32 heme oxygenase 1 Homo sapiens 93-97 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. Sulfhydryl Compounds 27-32 heme oxygenase 1 Homo sapiens 189-193 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. 15d-pgj 109-116 heme oxygenase 1 Homo sapiens 93-97 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. Sulfhydryl Compounds 143-148 heme oxygenase 1 Homo sapiens 189-193 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. Disulfides 149-158 heme oxygenase 1 Homo sapiens 93-97 14998722-10 2004 These results suggest that thiol antioxidant and reducing agents attenuate the expression of HO-1 induced by 15d-PGJ(2), and that the cellular thiol-disulfide redox status may be linked to HO-1 activation. Disulfides 149-158 heme oxygenase 1 Homo sapiens 189-193 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Iron 87-91 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Heme 54-58 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Heme 54-58 heme oxygenase 1 Homo sapiens 18-22 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 18-22 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. ammonium ferrous sulfate 81-85 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. ammonium ferrous sulfate 81-85 heme oxygenase 1 Homo sapiens 18-22 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Biliverdine 91-101 heme oxygenase 1 Homo sapiens 18-22 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Bilirubin 132-141 heme oxygenase 1 Homo sapiens 0-16 15004156-1 2004 Heme oxygenase-1 (HO-1) cleaves the porphyrin ring of heme into carbon monoxide, Fe2+, and biliverdin, which is then converted into bilirubin. Bilirubin 132-141 heme oxygenase 1 Homo sapiens 18-22 15004156-8 2004 Bilirubin and/or Fe2+ chelation mimicked the effects of HO-1, whereas biliverdin or carbon monoxide did not. Bilirubin 0-9 heme oxygenase 1 Homo sapiens 56-60 15004156-8 2004 Bilirubin and/or Fe2+ chelation mimicked the effects of HO-1, whereas biliverdin or carbon monoxide did not. ammonium ferrous sulfate 17-21 heme oxygenase 1 Homo sapiens 56-60 15004156-10 2004 This effect of HO-1 is mediated by bilirubin and/or by a decrease of free intracellular Fe2+ but probably not by biliverdin or carbon monoxide. Bilirubin 35-44 heme oxygenase 1 Homo sapiens 15-19 15004156-10 2004 This effect of HO-1 is mediated by bilirubin and/or by a decrease of free intracellular Fe2+ but probably not by biliverdin or carbon monoxide. ammonium ferrous sulfate 88-92 heme oxygenase 1 Homo sapiens 15-19 15028349-3 2004 A dinucleotide repeat in the HO-1 gene promoter shows a length polymorphism that modulates HO-1 gene transcription. Dinucleoside Phosphates 2-14 heme oxygenase 1 Homo sapiens 29-33 15028349-3 2004 A dinucleotide repeat in the HO-1 gene promoter shows a length polymorphism that modulates HO-1 gene transcription. Dinucleoside Phosphates 2-14 heme oxygenase 1 Homo sapiens 91-95 15028349-4 2004 Short (<25 guanosine thymidine [GT]) repeats are associated with a 10-fold greater up-regulation of HO-1 than are longer repeats. guanosine thymidine 14-33 heme oxygenase 1 Homo sapiens 103-107 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Iron 87-91 heme oxygenase 1 Homo sapiens 18-22 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 0-16 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 18-22 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 110-112 heme oxygenase 1 Homo sapiens 0-16 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Carbon Monoxide 110-112 heme oxygenase 1 Homo sapiens 18-22 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Biliverdine 119-129 heme oxygenase 1 Homo sapiens 0-16 14766239-1 2004 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation releasing iron, carbon monoxide (CO), and biliverdin. Biliverdine 119-129 heme oxygenase 1 Homo sapiens 18-22 14660625-11 2004 Heme oxygenase-1 expression was increased in rho(0) cells, and a heme oxygenase-1 inhibitor decreased the induction of MnSOD in rho(0) cells and their resistance against ROS donors. ros 170-173 heme oxygenase 1 Homo sapiens 0-16 15069378-3 2004 The HO enzyme exists in three isophorms: HO-1 is expressed at low levels under physiological conditions, but is induced by numerous factors, including oxidative stress, inflammation, nitric oxide, an elevated level of substrate, and hypoxia. Nitric Oxide 183-195 heme oxygenase 1 Homo sapiens 41-45 14761930-5 2004 Several stimuli implicated in the pathogenesis of renal injury, such as heme, nitric oxide, growth factors, angiotensin II, cytokines, and nephrotoxins, induce HO-1. Heme 72-76 heme oxygenase 1 Homo sapiens 160-164 14761930-5 2004 Several stimuli implicated in the pathogenesis of renal injury, such as heme, nitric oxide, growth factors, angiotensin II, cytokines, and nephrotoxins, induce HO-1. Nitric Oxide 78-90 heme oxygenase 1 Homo sapiens 160-164 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Iron 135-139 heme oxygenase 1 Homo sapiens 18-22 15105257-0 2004 Heme oxygenase-1: transducer of pathological brain iron sequestration under oxidative stress. Iron 51-55 heme oxygenase 1 Homo sapiens 0-16 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Heme 65-69 heme oxygenase 1 Homo sapiens 0-16 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Heme 65-69 heme oxygenase 1 Homo sapiens 18-22 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 0-16 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Biliverdine 73-83 heme oxygenase 1 Homo sapiens 18-22 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Iron 90-94 heme oxygenase 1 Homo sapiens 0-16 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Iron 90-94 heme oxygenase 1 Homo sapiens 18-22 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Carbon Monoxide 100-115 heme oxygenase 1 Homo sapiens 0-16 15105257-2 2004 Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that degrades heme to biliverdin, free iron, and carbon monoxide. Carbon Monoxide 100-115 heme oxygenase 1 Homo sapiens 18-22 15105257-4 2004 A model is presented implicating glial HO-1 induction as a "final common pathway" leading to pathological iron sequestration and mitochondrial insufficiency in a host of human CNS disorders. Iron 106-110 heme oxygenase 1 Homo sapiens 39-43 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Heme 85-89 heme oxygenase 1 Homo sapiens 0-16 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Heme 85-89 heme oxygenase 1 Homo sapiens 18-22 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Biliverdine 98-108 heme oxygenase 1 Homo sapiens 0-16 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Biliverdine 98-108 heme oxygenase 1 Homo sapiens 18-22 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 0-16 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 18-22 14735461-1 2004 Heme oxygenase-1 (HO-1), an inducible enzyme that catalyzes oxidative degradation of heme to form biliverdin, carbon monoxide and free iron, may protect tumor cells against oxidative stress, thus contributing to rapid tumor growth in vivo. Iron 135-139 heme oxygenase 1 Homo sapiens 0-16 14660625-11 2004 Heme oxygenase-1 expression was increased in rho(0) cells, and a heme oxygenase-1 inhibitor decreased the induction of MnSOD in rho(0) cells and their resistance against ROS donors. ros 170-173 heme oxygenase 1 Homo sapiens 65-81 14733911-0 2004 Antioxidant action of L-alanine: heme oxygenase-1 and ferritin as possible mediators. Alanine 22-31 heme oxygenase 1 Homo sapiens 33-49 15084931-2 2004 Inducibility of this enzyme is modulated by a (GT)n dinucleotide length polymorphism in the HO-1 gene promoter. Dinucleoside Phosphates 52-64 heme oxygenase 1 Homo sapiens 92-96 14733911-5 2004 A protective effect similar to L-alanine was observed when preincubating the cells with iron-free apoferritin or the antioxidant HO-1 product, bilirubin. Bilirubin 143-152 heme oxygenase 1 Homo sapiens 129-133 14733911-6 2004 The present study demonstrates that L-alanine stimulates expression of the antioxidant defense proteins HO-1 and ferritin in endothelial cells. Alanine 36-45 heme oxygenase 1 Homo sapiens 104-121 14715242-9 2004 We conclude that induction of HO-1 by 15d-PGJ(2) results in augmentation of VEGF synthesis in normoxia. 15d-pgj 38-45 heme oxygenase 1 Homo sapiens 30-34 14525760-9 2004 Furthermore, in the LPS-induced model of inflammatory angiogenesis, induction of HO-1 with cobalt protoporphyrin significantly inhibited leukocyte invasion into LPS-conditioned Matrigel and thus prevented the subsequent angiogenesis. cobaltiprotoporphyrin 91-112 heme oxygenase 1 Homo sapiens 81-85 14968347-7 2004 Additionally, PETriN enhanced the enzymatic activity of HO-1 measured as formation of the HO-1 metabolites bilirubin (Figure 3) and carbon monoxide (Figure 4) in lysates from endothelial cells. pentrinitrol 14-20 heme oxygenase 1 Homo sapiens 56-60 14968347-7 2004 Additionally, PETriN enhanced the enzymatic activity of HO-1 measured as formation of the HO-1 metabolites bilirubin (Figure 3) and carbon monoxide (Figure 4) in lysates from endothelial cells. pentrinitrol 14-20 heme oxygenase 1 Homo sapiens 90-94 14968347-7 2004 Additionally, PETriN enhanced the enzymatic activity of HO-1 measured as formation of the HO-1 metabolites bilirubin (Figure 3) and carbon monoxide (Figure 4) in lysates from endothelial cells. Bilirubin 107-116 heme oxygenase 1 Homo sapiens 56-60 14968347-7 2004 Additionally, PETriN enhanced the enzymatic activity of HO-1 measured as formation of the HO-1 metabolites bilirubin (Figure 3) and carbon monoxide (Figure 4) in lysates from endothelial cells. Bilirubin 107-116 heme oxygenase 1 Homo sapiens 90-94 14968347-7 2004 Additionally, PETriN enhanced the enzymatic activity of HO-1 measured as formation of the HO-1 metabolites bilirubin (Figure 3) and carbon monoxide (Figure 4) in lysates from endothelial cells. Carbon Monoxide 132-147 heme oxygenase 1 Homo sapiens 56-60 14968347-8 2004 HO-1 induction subsequently led to a marked increase in protein expression of a second antioxidant protein, ferritin, via the HO-1-dependent release of free iron from endogenous heme sources (Figures 1 and 5). Iron 157-161 heme oxygenase 1 Homo sapiens 0-4 14968347-8 2004 HO-1 induction subsequently led to a marked increase in protein expression of a second antioxidant protein, ferritin, via the HO-1-dependent release of free iron from endogenous heme sources (Figures 1 and 5). Iron 157-161 heme oxygenase 1 Homo sapiens 126-130 14968347-8 2004 HO-1 induction subsequently led to a marked increase in protein expression of a second antioxidant protein, ferritin, via the HO-1-dependent release of free iron from endogenous heme sources (Figures 1 and 5). Heme 178-182 heme oxygenase 1 Homo sapiens 0-4 14968347-8 2004 HO-1 induction subsequently led to a marked increase in protein expression of a second antioxidant protein, ferritin, via the HO-1-dependent release of free iron from endogenous heme sources (Figures 1 and 5). Heme 178-182 heme oxygenase 1 Homo sapiens 126-130 14968347-19 2004 Thus, the ability to activate HO-1 induction and associated antioxidant pathways apparently distinguishes PETN from other long-acting nitrates and may explain their different patterns of action in vivo (Figure 9). Pentaerythritol Tetranitrate 106-110 heme oxygenase 1 Homo sapiens 30-34 14968347-19 2004 Thus, the ability to activate HO-1 induction and associated antioxidant pathways apparently distinguishes PETN from other long-acting nitrates and may explain their different patterns of action in vivo (Figure 9). Nitrates 134-142 heme oxygenase 1 Homo sapiens 30-34 14707100-2 2004 We have recently shown that carbon monoxide (CO), an enzymatic product of heme oxygenase-1 (HO-1), confers potent antiproliferative effects in airway and vascular smooth muscle cells. Carbon Monoxide 28-43 heme oxygenase 1 Homo sapiens 74-90 14707100-2 2004 We have recently shown that carbon monoxide (CO), an enzymatic product of heme oxygenase-1 (HO-1), confers potent antiproliferative effects in airway and vascular smooth muscle cells. Carbon Monoxide 45-47 heme oxygenase 1 Homo sapiens 74-90 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Heme 100-104 heme oxygenase 1 Homo sapiens 14-30 14647439-4 2004 The cells were treated with hemin and cadmium to induce HO-1. Hemin 28-33 heme oxygenase 1 Homo sapiens 56-60 14647439-4 2004 The cells were treated with hemin and cadmium to induce HO-1. Cadmium 38-45 heme oxygenase 1 Homo sapiens 56-60 14647439-5 2004 The result showed that HO-1 protein was significantly induced by hemin and cadmium in both cells tested. Hemin 65-70 heme oxygenase 1 Homo sapiens 23-27 14647439-5 2004 The result showed that HO-1 protein was significantly induced by hemin and cadmium in both cells tested. Cadmium 75-82 heme oxygenase 1 Homo sapiens 23-27 14647439-8 2004 The levels of HO-1 and p21 induced were significantly inhibited by p38 mitogen-activated protein kinase (p38 MAPK) inhibitor (SB203580) and extracellular-regulated kinase (ERK) inhibitor (PD098059). SB 203580 126-134 heme oxygenase 1 Homo sapiens 14-18 14647439-8 2004 The levels of HO-1 and p21 induced were significantly inhibited by p38 mitogen-activated protein kinase (p38 MAPK) inhibitor (SB203580) and extracellular-regulated kinase (ERK) inhibitor (PD098059). 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 188-196 heme oxygenase 1 Homo sapiens 14-18 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Heme 100-104 heme oxygenase 1 Homo sapiens 32-36 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Biliverdine 110-123 heme oxygenase 1 Homo sapiens 14-30 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Biliverdine 110-123 heme oxygenase 1 Homo sapiens 32-36 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 14-30 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 32-36 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Iron 159-163 heme oxygenase 1 Homo sapiens 14-30 14960194-1 2004 INTRODUCTION: Heme oxygenase-1 (HO-1) is a stress response enzyme, which catalyses the breakdown of heme into biliverdin-IX alpha, carbon monoxide and ferrous iron. Iron 159-163 heme oxygenase 1 Homo sapiens 32-36 14960194-4 2004 RESULTS: HO-1 induction with cobalt protoporphorin (Co-PP) dose-dependently protected against apoptotic cell death as well as neutrophil-mediated oncosis in the galactosamine/endotoxin (Gal/ET) shock model. cobalt protoporphorin 29-50 heme oxygenase 1 Homo sapiens 9-13 14960194-4 2004 RESULTS: HO-1 induction with cobalt protoporphorin (Co-PP) dose-dependently protected against apoptotic cell death as well as neutrophil-mediated oncosis in the galactosamine/endotoxin (Gal/ET) shock model. co-pp 52-57 heme oxygenase 1 Homo sapiens 9-13 14960194-4 2004 RESULTS: HO-1 induction with cobalt protoporphorin (Co-PP) dose-dependently protected against apoptotic cell death as well as neutrophil-mediated oncosis in the galactosamine/endotoxin (Gal/ET) shock model. Galactosamine 161-174 heme oxygenase 1 Homo sapiens 9-13 14960194-5 2004 Induction of HO-1 with Co-PP dose-dependently protected against neutrophil-mediated oncosis as indicated by attenuated ALT release and TNF-mediated apoptotic cell death as indicated by reduced caspase-3 activation. co-pp 23-28 heme oxygenase 1 Homo sapiens 13-17 14683741-11 2004 These findings suggest the genetic contribution to stent restenosis and support the notion that the long dinucleotide GT repeat in promotor region may interfere with HO-1 gene transcription, leading to decreased vascular protection upon injury. Dinucleoside Phosphates 105-117 heme oxygenase 1 Homo sapiens 166-170 15777021-6 2004 The existence of a constitutive haem oxygenase (HO-2), mainly present in the vasculature and nervous system, and an inducible haem oxygenase (HO-1), which is highly expressed during stress conditions in all tissues, also suggests that cells have evolved a fine control of this enzymic pathway to ultimately regulate haem consumption and to ensure production of CO, biliverdin/bilirubin and iron during physiological and pathophysiological situations. Carbon Monoxide 361-363 heme oxygenase 1 Homo sapiens 142-146 15777021-6 2004 The existence of a constitutive haem oxygenase (HO-2), mainly present in the vasculature and nervous system, and an inducible haem oxygenase (HO-1), which is highly expressed during stress conditions in all tissues, also suggests that cells have evolved a fine control of this enzymic pathway to ultimately regulate haem consumption and to ensure production of CO, biliverdin/bilirubin and iron during physiological and pathophysiological situations. Biliverdine 365-375 heme oxygenase 1 Homo sapiens 142-146 15777021-6 2004 The existence of a constitutive haem oxygenase (HO-2), mainly present in the vasculature and nervous system, and an inducible haem oxygenase (HO-1), which is highly expressed during stress conditions in all tissues, also suggests that cells have evolved a fine control of this enzymic pathway to ultimately regulate haem consumption and to ensure production of CO, biliverdin/bilirubin and iron during physiological and pathophysiological situations. Bilirubin 376-385 heme oxygenase 1 Homo sapiens 142-146 15777021-6 2004 The existence of a constitutive haem oxygenase (HO-2), mainly present in the vasculature and nervous system, and an inducible haem oxygenase (HO-1), which is highly expressed during stress conditions in all tissues, also suggests that cells have evolved a fine control of this enzymic pathway to ultimately regulate haem consumption and to ensure production of CO, biliverdin/bilirubin and iron during physiological and pathophysiological situations. Iron 390-394 heme oxygenase 1 Homo sapiens 142-146 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Heme 59-63 heme oxygenase 1 Homo sapiens 6-22 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Heme 59-63 heme oxygenase 1 Homo sapiens 24-28 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Iron 111-115 heme oxygenase 1 Homo sapiens 6-22 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Iron 111-115 heme oxygenase 1 Homo sapiens 24-28 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Biliverdine 117-127 heme oxygenase 1 Homo sapiens 6-22 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Biliverdine 117-127 heme oxygenase 1 Homo sapiens 24-28 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 133-148 heme oxygenase 1 Homo sapiens 6-22 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 133-148 heme oxygenase 1 Homo sapiens 24-28 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 150-152 heme oxygenase 1 Homo sapiens 6-22 14683741-1 2004 AIMS: Heme oxygenase-1 (HO-1) is a rate-limiting enzyme in heme degradation, leading to the generation of free iron, biliverdin, and carbon monoxide (CO). Carbon Monoxide 150-152 heme oxygenase 1 Homo sapiens 24-28 14683741-3 2004 A dinucleotide GT repeat in the promotor region of human HO-1 gene shows a length polymorphism that modulates the level of gene transcription. Dinucleoside Phosphates 2-14 heme oxygenase 1 Homo sapiens 57-61 14523007-0 2003 Oxidized phospholipids induce expression of human heme oxygenase-1 involving activation of cAMP-responsive element-binding protein. Phospholipids 9-22 heme oxygenase 1 Homo sapiens 50-66 14702110-7 2004 Avicins inhibited epidermal hyperplasia, reduced p53 mutation, enhanced apoptosis, decreased generation of 8-hydroxy-2"-deoxyguanosine, and enhanced expression of NADPH:quinone oxidoreductase 1 and heme oxygenase-1. avicins 0-7 heme oxygenase 1 Homo sapiens 198-214 15140586-2 2004 A (GT)n dinucleotide repeat polymorphism in the HO-1 promoter has been shown to modulate HO-1 gene expression. Dinucleoside Phosphates 8-20 heme oxygenase 1 Homo sapiens 48-52 15140586-2 2004 A (GT)n dinucleotide repeat polymorphism in the HO-1 promoter has been shown to modulate HO-1 gene expression. Dinucleoside Phosphates 8-20 heme oxygenase 1 Homo sapiens 89-93 14691581-0 2004 A microsatellite polymorphism in the heme oxygenase-1 gene promoter is associated with increased bilirubin and HDL levels but not with coronary artery disease. Bilirubin 97-106 heme oxygenase 1 Homo sapiens 37-53 14691581-1 2004 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous anti-oxidant bilirubin which exerts beneficial effects against arteriosclerosis. Bilirubin 85-94 heme oxygenase 1 Homo sapiens 0-16 14691581-1 2004 Heme oxygenase 1 (HO-1) is involved in the generation of the endogenous anti-oxidant bilirubin which exerts beneficial effects against arteriosclerosis. Bilirubin 85-94 heme oxygenase 1 Homo sapiens 18-22 14523007-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, protects against oxidative stress, and shows potent anti-inflammatory effects. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 14523007-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, protects against oxidative stress, and shows potent anti-inflammatory effects. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 14523007-2 2003 Oxidized phospholipids, which are generated during inflammation and apoptosis, modulate the inflammatory response by inducing the expression of several genes including HO-1. Phospholipids 9-22 heme oxygenase 1 Homo sapiens 168-172 14523007-3 2003 Here we investigated the signaling pathways and transcriptional events involved in the induction of HO-1 gene expression by oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) in human umbilical vein endothelial cells. 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine 133-190 heme oxygenase 1 Homo sapiens 100-104 14523007-3 2003 Here we investigated the signaling pathways and transcriptional events involved in the induction of HO-1 gene expression by oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) in human umbilical vein endothelial cells. oxpapc 192-198 heme oxygenase 1 Homo sapiens 100-104 14523007-7 2003 We identified a cAMP-responsive element and a Maf recognition element to be involved in the transcriptional activation of the HO-1 promoter by OxPAPC. Cyclic AMP 16-20 heme oxygenase 1 Homo sapiens 126-130 14644359-5 2003 The expression of hsp 32 and hsp70 was studied in human diploid lung fibroblasts (HEL cells) and human monocytic leukaemia cells (THP-1 cells) incubated in vitro with different concentrations of dibenzo[a,l]pyrene (DB[a,l]P), 1-nitropyrene, (NP), 4-aminobiphenyl (ABP), ACN and EOM for different periods of time. dibenzo(a,l)pyrene 195-213 heme oxygenase 1 Homo sapiens 18-24 14635185-1 2003 The purpose of the present study was to examine the role of human heme oxygenase (human HO-1) in cell cycle progression following exposure to heme or human HO-1 gene transfer and to identify target genes associated with human HO-1-meditated increases in cell cycle progression using cDNA microarray technology. Heme 66-70 heme oxygenase 1 Homo sapiens 88-92 14635185-1 2003 The purpose of the present study was to examine the role of human heme oxygenase (human HO-1) in cell cycle progression following exposure to heme or human HO-1 gene transfer and to identify target genes associated with human HO-1-meditated increases in cell cycle progression using cDNA microarray technology. Heme 66-70 heme oxygenase 1 Homo sapiens 156-160 14635185-1 2003 The purpose of the present study was to examine the role of human heme oxygenase (human HO-1) in cell cycle progression following exposure to heme or human HO-1 gene transfer and to identify target genes associated with human HO-1-meditated increases in cell cycle progression using cDNA microarray technology. Heme 66-70 heme oxygenase 1 Homo sapiens 156-160 14635185-2 2003 Heme-induced robust human HO-1 expression in quiescent human microvessel endothelial cells cultured in 1% FBS and the levels of human HO-1 expression progressively declined without a change in the cell cyclin. Heme 0-4 heme oxygenase 1 Homo sapiens 26-30 14635185-2 2003 Heme-induced robust human HO-1 expression in quiescent human microvessel endothelial cells cultured in 1% FBS and the levels of human HO-1 expression progressively declined without a change in the cell cyclin. Heme 0-4 heme oxygenase 1 Homo sapiens 134-138 14637166-0 2003 Gastroprotection by vitamin C--a heme oxygenase-1-dependent mechanism? Ascorbic Acid 20-29 heme oxygenase 1 Homo sapiens 33-49 14637166-5 2003 HO-1 mRNA was significantly elevated by either ASA or vitamin C in gastric epithelial cells, combination of both substances further increased expression. Aspirin 47-50 heme oxygenase 1 Homo sapiens 0-4 14637166-5 2003 HO-1 mRNA was significantly elevated by either ASA or vitamin C in gastric epithelial cells, combination of both substances further increased expression. Ascorbic Acid 54-63 heme oxygenase 1 Homo sapiens 0-4 14637166-6 2003 HO-1 protein and enzyme activity rose in cells exposed to vitamin C alone or combined with ASA, but not after stimulation with ASA alone. Ascorbic Acid 58-67 heme oxygenase 1 Homo sapiens 0-4 14637166-6 2003 HO-1 protein and enzyme activity rose in cells exposed to vitamin C alone or combined with ASA, but not after stimulation with ASA alone. Aspirin 91-94 heme oxygenase 1 Homo sapiens 0-4 14637166-6 2003 HO-1 protein and enzyme activity rose in cells exposed to vitamin C alone or combined with ASA, but not after stimulation with ASA alone. Aspirin 127-130 heme oxygenase 1 Homo sapiens 0-4 14637166-7 2003 In contrast to endothelia, in which ASA simultaneously induces HO-1 mRNA and protein expression, gastric epithelial cells require vitamin C to translate HO-1 mRNA into active protein, which then may exert gastroprotection by its antioxidant and vasodilative properties. Aspirin 36-39 heme oxygenase 1 Homo sapiens 63-67 14637166-7 2003 In contrast to endothelia, in which ASA simultaneously induces HO-1 mRNA and protein expression, gastric epithelial cells require vitamin C to translate HO-1 mRNA into active protein, which then may exert gastroprotection by its antioxidant and vasodilative properties. Ascorbic Acid 130-139 heme oxygenase 1 Homo sapiens 153-157 14644359-7 2003 ACN induced the expression of hsp 32 as well as hsp70 in HEL and THP-1 cells, which probably reflects its ability to induce oxidative stress. Acrylonitrile 0-3 heme oxygenase 1 Homo sapiens 30-36 14615405-6 2003 Preexposure (24 h) in a combination of low O2 and low glucose concentrations decreased the protein content of the HO-1 isoform by 59.6% (P < 0.05), whereas preexposure (24 h) to low glucose concentration alone increased HO-2 content by 28.2% in chorionic villi explants (P < 0.05). Oxygen 43-45 heme oxygenase 1 Homo sapiens 114-118 14504288-3 2003 The small Maf proteins appear to be critical regulators of heme oxygenase (HO)-1, an anti-oxidant defense enzyme that degrades heme into iron, carbon monoxide, and biliverdin. Iron 137-141 heme oxygenase 1 Homo sapiens 59-80 14504288-3 2003 The small Maf proteins appear to be critical regulators of heme oxygenase (HO)-1, an anti-oxidant defense enzyme that degrades heme into iron, carbon monoxide, and biliverdin. Carbon Monoxide 143-158 heme oxygenase 1 Homo sapiens 59-80 14504288-3 2003 The small Maf proteins appear to be critical regulators of heme oxygenase (HO)-1, an anti-oxidant defense enzyme that degrades heme into iron, carbon monoxide, and biliverdin. Biliverdine 164-174 heme oxygenase 1 Homo sapiens 59-80 14615405-6 2003 Preexposure (24 h) in a combination of low O2 and low glucose concentrations decreased the protein content of the HO-1 isoform by 59.6% (P < 0.05), whereas preexposure (24 h) to low glucose concentration alone increased HO-2 content by 28.2% in chorionic villi explants (P < 0.05). Glucose 54-61 heme oxygenase 1 Homo sapiens 114-118 14615405-6 2003 Preexposure (24 h) in a combination of low O2 and low glucose concentrations decreased the protein content of the HO-1 isoform by 59.6% (P < 0.05), whereas preexposure (24 h) to low glucose concentration alone increased HO-2 content by 28.2% in chorionic villi explants (P < 0.05). Glucose 185-192 heme oxygenase 1 Homo sapiens 114-118 14638927-9 2003 Renal function at the first week posttransplantation (analyzed by serum creatinine levels) showed a significant correlation with both HO-1 and VEGF mRNA expression, reinforcing the protective role of both genes in the early events of transplantation. Creatinine 72-82 heme oxygenase 1 Homo sapiens 134-138 12907459-8 2003 Comparative Northern analyses of iron-related genes after erythrophagocytosis revealed a 16-fold increase in FPN1 levels after 3 hours, a 10-fold increase in heme oxygenase-1 (HO-1) after 3 hours, a 2-fold increase in natural resistance macrophage-associated protein 1 (Nramp1) levels after 6 hours, but no change in divalent metal ion transporter 1 (DMT1) levels. Iron 33-37 heme oxygenase 1 Homo sapiens 158-174 14522998-9 2003 Heme oxygenase-1 was induced at low extract dose and with minimal decline in the GSH/GSSG ratio, whereas MAP kinase activation required a higher chemical dose and incremental levels of oxidative stress. Glutathione Disulfide 81-84 heme oxygenase 1 Homo sapiens 0-16 14598260-4 2003 Our aim was to explore the role of the heme oxygenase-1/carbon monoxide pathway in the pathogenesis of experimental hepatopulmonary syndrome. Carbon Monoxide 56-71 heme oxygenase 1 Homo sapiens 39-55 14562166-1 2003 INTRODUCTION: Heme oxygenase (HO) isoforms, HO-1, and HO-2, are responsible for heme breakdown to iron and carbon monoxide (CO). Heme 80-84 heme oxygenase 1 Homo sapiens 44-48 14562166-1 2003 INTRODUCTION: Heme oxygenase (HO) isoforms, HO-1, and HO-2, are responsible for heme breakdown to iron and carbon monoxide (CO). Iron 98-102 heme oxygenase 1 Homo sapiens 44-48 14562166-1 2003 INTRODUCTION: Heme oxygenase (HO) isoforms, HO-1, and HO-2, are responsible for heme breakdown to iron and carbon monoxide (CO). Carbon Monoxide 107-122 heme oxygenase 1 Homo sapiens 44-48 14624702-1 2003 BACKGROUND: As well as being inducible by haem, haemoxygenase -1 (HO-1) is also induced by interleukin-10 and an anti-inflammatory prostaglandin, 15d PGJ2, the carbon monoxide thus produced mediating the anti-inflammatory effects of these molecules. Prostaglandins 131-144 heme oxygenase 1 Homo sapiens 48-70 14624702-1 2003 BACKGROUND: As well as being inducible by haem, haemoxygenase -1 (HO-1) is also induced by interleukin-10 and an anti-inflammatory prostaglandin, 15d PGJ2, the carbon monoxide thus produced mediating the anti-inflammatory effects of these molecules. 9-deoxy-delta-9-prostaglandin D2 150-154 heme oxygenase 1 Homo sapiens 48-70 14624702-1 2003 BACKGROUND: As well as being inducible by haem, haemoxygenase -1 (HO-1) is also induced by interleukin-10 and an anti-inflammatory prostaglandin, 15d PGJ2, the carbon monoxide thus produced mediating the anti-inflammatory effects of these molecules. Carbon Monoxide 160-175 heme oxygenase 1 Homo sapiens 48-70 14562166-1 2003 INTRODUCTION: Heme oxygenase (HO) isoforms, HO-1, and HO-2, are responsible for heme breakdown to iron and carbon monoxide (CO). Carbon Monoxide 124-126 heme oxygenase 1 Homo sapiens 44-48 14598260-10 2003 CONCLUSIONS: The heme oxygenase-1/carbon monoxide system is an important contributor to the progression of experimental hepatopulmonary syndrome in addition to alterations in the endothelial nitric oxide synthase/nitric oxide pathway. Carbon Monoxide 34-49 heme oxygenase 1 Homo sapiens 17-33 14696395-1 2003 BACKGROUND/AIMS: Hemeoxygenase-1 produces carbon monoxide as a byproduct of hemoglobin metabolism. Carbon Monoxide 42-57 heme oxygenase 1 Homo sapiens 17-32 14614191-6 2003 Heme oxygenase-1 (HO-1) protein level is increased 24 h after HBO exposure and inhibition of HO-1 activity by tin-mesoporphyrin increased HBO genotoxicity. tin mesoporphyrin 110-127 heme oxygenase 1 Homo sapiens 0-16 12882997-3 2003 Using antisense oligonucleotides, we achieved a block of iNOS protein formation, accompanied by a strong decrease in the expression of the protective stress response genes bcl-2, vascular endothelial growth factor, and heme oxygenase-1 (HO-1). Oligonucleotides 16-32 heme oxygenase 1 Homo sapiens 219-235 14614191-6 2003 Heme oxygenase-1 (HO-1) protein level is increased 24 h after HBO exposure and inhibition of HO-1 activity by tin-mesoporphyrin increased HBO genotoxicity. tin mesoporphyrin 110-127 heme oxygenase 1 Homo sapiens 93-97 12969148-6 2003 RESULTS: In HRPTECs, both HO-1 mRNA expression and protein production peaked at around 12 h and persisted until 24 h after hemin stimulation. Hemin 123-128 heme oxygenase 1 Homo sapiens 26-30 12969148-10 2003 Tin protoporphyrin (SnPP), an inhibitor of HO function, significantly reversed the cytoprotection by HO-1. tin protoporphyrin IX 0-18 heme oxygenase 1 Homo sapiens 101-105 12969148-10 2003 Tin protoporphyrin (SnPP), an inhibitor of HO function, significantly reversed the cytoprotection by HO-1. S-Nitroso-N-propionyl-D,L-penicillamine 20-24 heme oxygenase 1 Homo sapiens 101-105 12933701-0 2003 Heme oxygenase-1 attenuates glucose-mediated cell growth arrest and apoptosis in human microvessel endothelial cells. Glucose 28-35 heme oxygenase 1 Homo sapiens 0-16 12933701-8 2003 These findings identify a novel effect of HO-1 on endothelial cell growth and indicate that heme metabolism and HO-1 expression regulate signaling systems in cells exposed to high glucose, which controls cell-cycle progression. Glucose 180-187 heme oxygenase 1 Homo sapiens 42-46 12933701-1 2003 Heme oxygenase-1 (HO-1) is a stress protein that has been suggested to participate in defense mechanisms against agents that may induce oxidative injury, such as heme and inflammatory molecules. Heme 162-166 heme oxygenase 1 Homo sapiens 0-16 12933701-8 2003 These findings identify a novel effect of HO-1 on endothelial cell growth and indicate that heme metabolism and HO-1 expression regulate signaling systems in cells exposed to high glucose, which controls cell-cycle progression. Glucose 180-187 heme oxygenase 1 Homo sapiens 112-116 12933701-1 2003 Heme oxygenase-1 (HO-1) is a stress protein that has been suggested to participate in defense mechanisms against agents that may induce oxidative injury, such as heme and inflammatory molecules. Heme 162-166 heme oxygenase 1 Homo sapiens 18-22 12933701-2 2003 Incubation of endothelial cells in a high-glucose (33 mmol/L) medium for 7 days resulted in a decrease of HO activity by 34% and a decrease in HO-1 and HO-2 proteins compared with cells exposed to low glucose (5 mmol/L) (P<0.05) or cells exposed to mannitol (33 mmol/L). Glucose 42-49 heme oxygenase 1 Homo sapiens 143-156 12933701-3 2003 Overexpression of HO-1 was coupled with an increase in HO activity and carbon monoxide synthesis, decreased cellular heme, and acceleration in all phases of the cell cycle (P<0.001). Carbon Monoxide 71-86 heme oxygenase 1 Homo sapiens 18-22 12933701-3 2003 Overexpression of HO-1 was coupled with an increase in HO activity and carbon monoxide synthesis, decreased cellular heme, and acceleration in all phases of the cell cycle (P<0.001). Heme 117-121 heme oxygenase 1 Homo sapiens 18-22 12933701-5 2003 Exposure to high glucose significantly decreased cell-cycle progression in control cells and in cells underexpressing HO-1 but did not decrease cell-cycle progression in cells overexpressing HO-1. Glucose 17-24 heme oxygenase 1 Homo sapiens 118-122 12933701-7 2003 The addition of tin-mesoporphyrin (SnMP), an inhibitor of HO activity, reversed the HO-1-mediated decrease of p21 and p27 in cells overexpressing HO-1. tin mesoporphyrin 16-33 heme oxygenase 1 Homo sapiens 84-88 12933701-7 2003 The addition of tin-mesoporphyrin (SnMP), an inhibitor of HO activity, reversed the HO-1-mediated decrease of p21 and p27 in cells overexpressing HO-1. tin mesoporphyrin 16-33 heme oxygenase 1 Homo sapiens 146-150 12933701-7 2003 The addition of tin-mesoporphyrin (SnMP), an inhibitor of HO activity, reversed the HO-1-mediated decrease of p21 and p27 in cells overexpressing HO-1. tin mesoporphyrin 35-39 heme oxygenase 1 Homo sapiens 84-88 12933701-7 2003 The addition of tin-mesoporphyrin (SnMP), an inhibitor of HO activity, reversed the HO-1-mediated decrease of p21 and p27 in cells overexpressing HO-1. tin mesoporphyrin 35-39 heme oxygenase 1 Homo sapiens 146-150 12933701-8 2003 These findings identify a novel effect of HO-1 on endothelial cell growth and indicate that heme metabolism and HO-1 expression regulate signaling systems in cells exposed to high glucose, which controls cell-cycle progression. Heme 92-96 heme oxygenase 1 Homo sapiens 42-46 12927812-2 2003 In cultured endothelial cells derived from human umbilical vein, aspirin (30-300 microM) increased heme oxygenase-1 (HO-1) protein levels in a concentration-dependent fashion up to fivefold over basal levels. Aspirin 65-72 heme oxygenase 1 Homo sapiens 99-115 12819228-11 2003 Although the active site residues differ between nm-HO and human HO-1, the close similarity in the H-bonded water network suggests a common mechanism shared by all heme oxygenases. Water 108-113 heme oxygenase 1 Homo sapiens 65-69 12927811-0 2003 Changes in temperature modulate heme oxygenase-1 induction by curcumin in renal epithelial cells. Curcumin 62-70 heme oxygenase 1 Homo sapiens 32-48 12927812-2 2003 In cultured endothelial cells derived from human umbilical vein, aspirin (30-300 microM) increased heme oxygenase-1 (HO-1) protein levels in a concentration-dependent fashion up to fivefold over basal levels. Aspirin 65-72 heme oxygenase 1 Homo sapiens 117-121 12927812-0 2003 Heme oxygenase-1 induction may explain the antioxidant profile of aspirin. Aspirin 66-73 heme oxygenase 1 Homo sapiens 0-16 12927812-3 2003 HO-1 induction was accompanied by a marked increase in catalytic activity of the enzyme as reflected by enhanced formation of both carbon monoxide and bilirubin. Carbon Monoxide 131-146 heme oxygenase 1 Homo sapiens 0-4 12927812-3 2003 HO-1 induction was accompanied by a marked increase in catalytic activity of the enzyme as reflected by enhanced formation of both carbon monoxide and bilirubin. Bilirubin 151-160 heme oxygenase 1 Homo sapiens 0-4 12927812-6 2003 The nitric oxide (NO) synthase blocker L-NAME prevented aspirin-dependent HO-1 induction. Nitric Oxide 4-16 heme oxygenase 1 Homo sapiens 74-78 12927812-6 2003 The nitric oxide (NO) synthase blocker L-NAME prevented aspirin-dependent HO-1 induction. NG-Nitroarginine Methyl Ester 39-45 heme oxygenase 1 Homo sapiens 74-78 12927812-6 2003 The nitric oxide (NO) synthase blocker L-NAME prevented aspirin-dependent HO-1 induction. Aspirin 56-63 heme oxygenase 1 Homo sapiens 74-78 12927812-7 2003 These findings demonstrate that aspirin targets HO-1, presumably via NO-dependent pathways. Aspirin 32-39 heme oxygenase 1 Homo sapiens 48-52 12927812-8 2003 Induction of HO-1 expression and activity may be a novel mechanism by which aspirin prevents cellular injury under inflammatory conditions and in cardiovascular disease. Aspirin 76-83 heme oxygenase 1 Homo sapiens 13-17 12927819-6 2003 Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). stannis mesoporphyrin 23-44 heme oxygenase 1 Homo sapiens 14-18 12927819-6 2003 Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). tin mesoporphyrin 46-50 heme oxygenase 1 Homo sapiens 14-18 12927819-6 2003 Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). 8-epi-isoprostane 110-127 heme oxygenase 1 Homo sapiens 14-18 12783778-0 2003 An internal enhancer regulates heme- and cadmium-mediated induction of human heme oxygenase-1. Heme 31-35 heme oxygenase 1 Homo sapiens 77-93 12783778-0 2003 An internal enhancer regulates heme- and cadmium-mediated induction of human heme oxygenase-1. Cadmium 41-48 heme oxygenase 1 Homo sapiens 77-93 12927819-6 2003 Inhibition of HO-1, by Stannis mesoporphyrin (SnMP), potentiated Ang II-mediated DNA damage and generation of 8-epi-isoprostane PGF(2 alpha). Prostaglandins F 128-131 heme oxygenase 1 Homo sapiens 14-18 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 12927819-9 2003 Upregulation of HO-1 ensures the generation of bilirubin and carbon monoxide (CO) in G(0)/G(1) phase to counteract Ang II-mediated oxidative DNA damage. Bilirubin 47-56 heme oxygenase 1 Homo sapiens 16-20 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Iron 88-92 heme oxygenase 1 Homo sapiens 0-16 12927819-9 2003 Upregulation of HO-1 ensures the generation of bilirubin and carbon monoxide (CO) in G(0)/G(1) phase to counteract Ang II-mediated oxidative DNA damage. Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 16-20 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Iron 88-92 heme oxygenase 1 Homo sapiens 18-22 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 0-16 12927819-9 2003 Upregulation of HO-1 ensures the generation of bilirubin and carbon monoxide (CO) in G(0)/G(1) phase to counteract Ang II-mediated oxidative DNA damage. Carbon Monoxide 78-80 heme oxygenase 1 Homo sapiens 16-20 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 18-22 12927819-10 2003 Inducibility of HO-1 in G(0)/G(1) phase is essential and probably regulated by a complex system involving oxygen species to assure controlled cell growth. Oxygen 106-112 heme oxygenase 1 Homo sapiens 16-20 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 0-16 12783778-1 2003 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 18-22 12941774-4 2003 Both basal HSP72 (r = 0.75, P < 0.001) and HO-1 (r = 0.50, P < 0.05) mRNA expression correlated with the glucose infusion rate during the clamp. Glucose 111-118 heme oxygenase 1 Homo sapiens 46-50 12783778-3 2003 The purpose of this study was to characterize the regulation of the human HO-1 gene in renal proximal tubule and aortic endothelial cells in response to heme and cadmium. Heme 153-157 heme oxygenase 1 Homo sapiens 74-78 12783778-3 2003 The purpose of this study was to characterize the regulation of the human HO-1 gene in renal proximal tubule and aortic endothelial cells in response to heme and cadmium. Cadmium 162-169 heme oxygenase 1 Homo sapiens 74-78 12783778-4 2003 Evaluation of multiple human HO-1 promoter-reporter constructs up to -9.1 kb demonstrated only a partial response to heme and cadmium. Heme 117-121 heme oxygenase 1 Homo sapiens 29-33 12783778-4 2003 Evaluation of multiple human HO-1 promoter-reporter constructs up to -9.1 kb demonstrated only a partial response to heme and cadmium. Cadmium 126-133 heme oxygenase 1 Homo sapiens 29-33 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Heme 131-135 heme oxygenase 1 Homo sapiens 62-66 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Heme 131-135 heme oxygenase 1 Homo sapiens 102-106 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Heme 131-135 heme oxygenase 1 Homo sapiens 102-106 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Cadmium 141-148 heme oxygenase 1 Homo sapiens 62-66 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Cadmium 141-148 heme oxygenase 1 Homo sapiens 102-106 12783778-7 2003 Our studies identified a novel enhancer internal to the human HO-1 gene that, in conjunction with the HO-1 promoter, recapitulates heme- and cadmium-mediated induction of the endogenous HO-1 gene. Cadmium 141-148 heme oxygenase 1 Homo sapiens 102-106 12925221-5 2003 Lomefloxacin also triggered various stress responses: heme-oxygenase-1 expression in fibroblasts, changes in p53 status as shown by the accumulation of p53 and p21 proteins or the induction of MDM2 and GADD45 genes, and stimulation of melanogenesis by increasing the tyrosinase activity in melanocytes. lomefloxacin 0-12 heme oxygenase 1 Homo sapiens 54-70 12958189-2 2003 HO-1 cleaves the heme porphyrin ring releasing Fe2+, which induces the expression of the Fe2+ sequestering protein ferritin. Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 12958189-2 2003 HO-1 cleaves the heme porphyrin ring releasing Fe2+, which induces the expression of the Fe2+ sequestering protein ferritin. ammonium ferrous sulfate 47-51 heme oxygenase 1 Homo sapiens 0-4 12958189-2 2003 HO-1 cleaves the heme porphyrin ring releasing Fe2+, which induces the expression of the Fe2+ sequestering protein ferritin. ammonium ferrous sulfate 89-93 heme oxygenase 1 Homo sapiens 0-4 12878207-1 2003 Vascular endothelial cells respond to nitric oxide by activating MAPK pathways and upregulating stress-activated proteins such as gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase-1 (HO-1). Nitric Oxide 38-50 heme oxygenase 1 Homo sapiens 180-196 12777398-8 2003 Increasing oxidative stress by the addition of arachidonic acid or depletion of glutathione further increased HO-1 induction. Arachidonic Acid 47-63 heme oxygenase 1 Homo sapiens 110-114 12777398-8 2003 Increasing oxidative stress by the addition of arachidonic acid or depletion of glutathione further increased HO-1 induction. Glutathione 80-91 heme oxygenase 1 Homo sapiens 110-114 12878207-1 2003 Vascular endothelial cells respond to nitric oxide by activating MAPK pathways and upregulating stress-activated proteins such as gamma-glutamylcysteine synthetase (gamma-GCS) and heme oxygenase-1 (HO-1). Nitric Oxide 38-50 heme oxygenase 1 Homo sapiens 198-202 12777398-10 2003 PD98059, a specific inhibitor of ERK MAPK, blocked the activity of a HO-1 reporter in E47 cells but not in C34 cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 heme oxygenase 1 Homo sapiens 69-73 12878207-7 2003 Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. Acetylcysteine 18-34 heme oxygenase 1 Homo sapiens 70-74 12878207-7 2003 Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 36-44 heme oxygenase 1 Homo sapiens 70-74 12878207-7 2003 Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. SB 203580 50-59 heme oxygenase 1 Homo sapiens 70-74 12878207-7 2003 Pretreatment with N-acetylcysteine, PD 98059, and SB 203580 decreased HO-1 upregulation in spermine NONOate-treated cells. spermine nitric oxide complex 91-107 heme oxygenase 1 Homo sapiens 70-74 12700136-6 2003 Induction of HO-1 gene expression by phorone, a glutathione depletor, and 4-hydroxy-2,3-nonenal (4-HNE), an end product of lipid peroxidation, was suppressed by a specific PKC inhibitor, Ro-31-8220, at concentrations that inhibit all isoforms in WI-38 cells. phorone 37-44 heme oxygenase 1 Homo sapiens 13-17 12700136-6 2003 Induction of HO-1 gene expression by phorone, a glutathione depletor, and 4-hydroxy-2,3-nonenal (4-HNE), an end product of lipid peroxidation, was suppressed by a specific PKC inhibitor, Ro-31-8220, at concentrations that inhibit all isoforms in WI-38 cells. Glutathione 48-59 heme oxygenase 1 Homo sapiens 13-17 12700136-6 2003 Induction of HO-1 gene expression by phorone, a glutathione depletor, and 4-hydroxy-2,3-nonenal (4-HNE), an end product of lipid peroxidation, was suppressed by a specific PKC inhibitor, Ro-31-8220, at concentrations that inhibit all isoforms in WI-38 cells. 4-hydroxy-2-nonenal 97-102 heme oxygenase 1 Homo sapiens 13-17 13678532-0 2003 Induction of heme oxygenase-1 in monocytes suppresses angiotensin II-elicited chemotactic activity through inhibition of CCR2: role of bilirubin and carbon monoxide generated by the enzyme. Bilirubin 135-144 heme oxygenase 1 Homo sapiens 13-29 13678532-0 2003 Induction of heme oxygenase-1 in monocytes suppresses angiotensin II-elicited chemotactic activity through inhibition of CCR2: role of bilirubin and carbon monoxide generated by the enzyme. Carbon Monoxide 149-164 heme oxygenase 1 Homo sapiens 13-29 13678532-2 2003 Heme oxygenase (HO) is a microsomal enzyme that catalyzes the degradation of heme into biliverdin, which is subsequently reduced to bilirubin, free iron, and carbon monoxide, and induction of HO-1 is potentially associated with cellular protection, especially against oxidative insults. Heme 77-81 heme oxygenase 1 Homo sapiens 192-196 13678532-2 2003 Heme oxygenase (HO) is a microsomal enzyme that catalyzes the degradation of heme into biliverdin, which is subsequently reduced to bilirubin, free iron, and carbon monoxide, and induction of HO-1 is potentially associated with cellular protection, especially against oxidative insults. Biliverdine 87-97 heme oxygenase 1 Homo sapiens 192-196 12872043-11 2003 As carbon monoxide, which is one of the products of HO-1, can attenuate nitric oxide-induced vasodilatation, a high expression of HO-1 may cause hypertension, especially in women. Carbon Monoxide 3-18 heme oxygenase 1 Homo sapiens 52-56 13678532-2 2003 Heme oxygenase (HO) is a microsomal enzyme that catalyzes the degradation of heme into biliverdin, which is subsequently reduced to bilirubin, free iron, and carbon monoxide, and induction of HO-1 is potentially associated with cellular protection, especially against oxidative insults. Bilirubin 132-141 heme oxygenase 1 Homo sapiens 192-196 13678532-2 2003 Heme oxygenase (HO) is a microsomal enzyme that catalyzes the degradation of heme into biliverdin, which is subsequently reduced to bilirubin, free iron, and carbon monoxide, and induction of HO-1 is potentially associated with cellular protection, especially against oxidative insults. Iron 148-152 heme oxygenase 1 Homo sapiens 192-196 13678532-2 2003 Heme oxygenase (HO) is a microsomal enzyme that catalyzes the degradation of heme into biliverdin, which is subsequently reduced to bilirubin, free iron, and carbon monoxide, and induction of HO-1 is potentially associated with cellular protection, especially against oxidative insults. Carbon Monoxide 158-173 heme oxygenase 1 Homo sapiens 192-196 13678532-6 2003 The induction of HO-1 in monocytes suppresses not only Ang II-stimulated superoxide formation, but also Ang II-enhanced chemotactic activity. Superoxides 73-83 heme oxygenase 1 Homo sapiens 17-21 13678532-7 2003 Exogenously applied bilirubin and carbon monoxide mimicked the inhibitory effect of HO-1 on the chemotactic response. Bilirubin 20-29 heme oxygenase 1 Homo sapiens 84-88 13678532-7 2003 Exogenously applied bilirubin and carbon monoxide mimicked the inhibitory effect of HO-1 on the chemotactic response. Carbon Monoxide 34-49 heme oxygenase 1 Homo sapiens 84-88 12891549-3 2003 The aim of the present study was to investigate the role of a stress-inducible protein, heme oxygenase-1 (HO-1), in the action of 15-d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 130-139 heme oxygenase 1 Homo sapiens 88-104 12891549-3 2003 The aim of the present study was to investigate the role of a stress-inducible protein, heme oxygenase-1 (HO-1), in the action of 15-d-PGJ2. 15-deoxy-delta(12,14)-prostaglandin J2 130-139 heme oxygenase 1 Homo sapiens 106-110 12891549-8 2003 HO-1 messenger RNA (mRNA) and protein also were detected in cultured human hepatic myofibroblasts and increased in response to 15-d-PGJ2 in a time- and dose-dependent manner. 15-deoxy-delta(12,14)-prostaglandin J2 127-136 heme oxygenase 1 Homo sapiens 0-4 12891549-9 2003 Induction of HO-1 in human hepatic myofibroblasts mediated 2 major antifibrogenic properties of 15-d-PGJ2, namely, inhibition of proliferation and of procollagen I mRNA expression. 15-deoxy-delta(12,14)-prostaglandin J2 96-105 heme oxygenase 1 Homo sapiens 13-17 12891549-10 2003 These effects were ascribed to bilirubin, one of the products of HO-1-mediated heme degradation. Bilirubin 31-40 heme oxygenase 1 Homo sapiens 65-69 12891549-10 2003 These effects were ascribed to bilirubin, one of the products of HO-1-mediated heme degradation. Heme 79-83 heme oxygenase 1 Homo sapiens 65-69 12805077-1 2003 OBJECTIVE: Several proatherogenic agents including oxidized LDL and its major component, 13-hydroperoxyoctadecadienoic acid (13-HPODE), upregulate heme oxygenase-1 (HO-1). 13-Hydroperoxyoctadecadienoic acid 89-123 heme oxygenase 1 Homo sapiens 147-163 12805077-1 2003 OBJECTIVE: Several proatherogenic agents including oxidized LDL and its major component, 13-hydroperoxyoctadecadienoic acid (13-HPODE), upregulate heme oxygenase-1 (HO-1). 13-Hydroperoxyoctadecadienoic acid 89-123 heme oxygenase 1 Homo sapiens 165-169 12805077-5 2003 Antioxidants such as N-acetylcysteine, iron chelation with deferoxamine mesylate, and protein kinase C inhibition with Go6976 blocked HO-1 induction. Go 6976 119-125 heme oxygenase 1 Homo sapiens 134-138 12872043-11 2003 As carbon monoxide, which is one of the products of HO-1, can attenuate nitric oxide-induced vasodilatation, a high expression of HO-1 may cause hypertension, especially in women. Carbon Monoxide 3-18 heme oxygenase 1 Homo sapiens 130-134 12872043-11 2003 As carbon monoxide, which is one of the products of HO-1, can attenuate nitric oxide-induced vasodilatation, a high expression of HO-1 may cause hypertension, especially in women. Nitric Oxide 72-84 heme oxygenase 1 Homo sapiens 52-56 12872043-11 2003 As carbon monoxide, which is one of the products of HO-1, can attenuate nitric oxide-induced vasodilatation, a high expression of HO-1 may cause hypertension, especially in women. Nitric Oxide 72-84 heme oxygenase 1 Homo sapiens 130-134 12909459-0 2003 Heme oxygenase-1: unleashing the protective properties of heme. Heme 58-62 heme oxygenase 1 Homo sapiens 0-16 12865654-8 2003 Pyrrolidine dithiocarbamate, an inhibitor of nuclear factor kappaB-dependent gene expression, dose dependently decreased HO-1 mRNA transcripts in human mononuclear cells subjected to oxidative stress while slightly increasing HO-1 gene expression in HepG2 cells. pyrrolidine dithiocarbamic acid 0-27 heme oxygenase 1 Homo sapiens 121-125 12865654-8 2003 Pyrrolidine dithiocarbamate, an inhibitor of nuclear factor kappaB-dependent gene expression, dose dependently decreased HO-1 mRNA transcripts in human mononuclear cells subjected to oxidative stress while slightly increasing HO-1 gene expression in HepG2 cells. pyrrolidine dithiocarbamic acid 0-27 heme oxygenase 1 Homo sapiens 226-230 12865654-9 2003 In contrast, HO-1 induction upon oxidative stress was attenuated in HepG2 cells by cycloheximide and dexamethasone. Cycloheximide 83-96 heme oxygenase 1 Homo sapiens 13-17 12865654-9 2003 In contrast, HO-1 induction upon oxidative stress was attenuated in HepG2 cells by cycloheximide and dexamethasone. Dexamethasone 101-114 heme oxygenase 1 Homo sapiens 13-17 12865654-12 2003 These data suggest a differential regulation of HO-1 gene expression in parenchymal and non-parenchymal human liver cells and may provide a topographic basis for the understanding of the role of the heme oxygenase/carbon monoxide pathway in human liver disease. Carbon Monoxide 214-229 heme oxygenase 1 Homo sapiens 48-52 12947311-5 2003 METHODS: Human umbilical vein endothelial cells were pretreated with hemin (100 micromol/L) for 5 hours, and the induction of HO-1 was confirmed by Western blot. Hemin 69-74 heme oxygenase 1 Homo sapiens 126-130 12947311-8 2003 RESULTS: HO-1 protein was increased 3-fold by exposure to hemin under all conditions. Hemin 58-63 heme oxygenase 1 Homo sapiens 9-13 12947311-11 2003 CONCLUSIONS: The induction of HO-1 by hemin results in inhibition of the proinflammatory response of endothelial cells, as evidenced by the inhibition of IL-8 production without affecting PGI(2) production. Epoprostenol 188-194 heme oxygenase 1 Homo sapiens 30-34 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Heme 34-38 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Heme 0-4 heme oxygenase 1 Homo sapiens 69-85 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Heme 0-4 heme oxygenase 1 Homo sapiens 87-91 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Carbon Monoxide 60-75 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Biliverdine 123-133 heme oxygenase 1 Homo sapiens 69-85 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Biliverdine 123-133 heme oxygenase 1 Homo sapiens 87-91 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Carbon Monoxide 77-79 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Bilirubin 134-143 heme oxygenase 1 Homo sapiens 69-85 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Bilirubin 134-143 heme oxygenase 1 Homo sapiens 87-91 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Biliverdine 82-92 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Carbon Monoxide 145-147 heme oxygenase 1 Homo sapiens 69-85 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Bilirubin 124-133 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Carbon Monoxide 145-147 heme oxygenase 1 Homo sapiens 87-91 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Iron 153-157 heme oxygenase 1 Homo sapiens 69-85 12909459-1 2003 Heme oxygenase (HO)-1 catabolizes heme into three products: carbon monoxide (CO), biliverdin (which is rapidly converted to bilirubin) and free iron (which leads to the induction of ferritin, an iron-binding protein). Iron 144-148 heme oxygenase 1 Homo sapiens 0-21 14580148-2 2003 Heme degradation is catalyzed by the two isozymes of heme oxygenase, heme oxygenase-1 (HO-1) and HO-2, eventually yielding biliverdin/bilirubin, CO, and iron. Iron 153-157 heme oxygenase 1 Homo sapiens 87-91 12909459-4 2003 We suggest that the products of HO-1 action could be valuable therapeutic agents and speculate that HO-1 functions as a "therapeutic funnel", mediating the beneficial effects attributed to other molecules, such as interleukin-10 (IL-10), inducible nitric oxide synthase (NOS2; iNOS) and prostaglandins. Prostaglandins 287-301 heme oxygenase 1 Homo sapiens 32-36 14580148-7 2003 Notably, HO-1 expression is induced by heme in all mammalian cells examined, but is repressed by hypoxia in certain types of cultured human cells. Heme 39-43 heme oxygenase 1 Homo sapiens 9-13 14580148-8 2003 The recent discovery of Bach1 as a heme-regulated and hypoxia-inducible repressor for transcription of the HO-1 gene has provided a missing link in the feedback control of heme catabolism. Heme 35-39 heme oxygenase 1 Homo sapiens 107-111 12909459-4 2003 We suggest that the products of HO-1 action could be valuable therapeutic agents and speculate that HO-1 functions as a "therapeutic funnel", mediating the beneficial effects attributed to other molecules, such as interleukin-10 (IL-10), inducible nitric oxide synthase (NOS2; iNOS) and prostaglandins. Prostaglandins 287-301 heme oxygenase 1 Homo sapiens 100-104 12832044-0 2003 Heme oxygenase-1 induction by endogenous nitric oxide: influence of intracellular glutathione. Nitric Oxide 41-53 heme oxygenase 1 Homo sapiens 0-16 12832044-0 2003 Heme oxygenase-1 induction by endogenous nitric oxide: influence of intracellular glutathione. Glutathione 82-93 heme oxygenase 1 Homo sapiens 0-16 12832044-2 2003 Protein levels of oxidative stress-sensitive heme oxygenase-1 (HO-1) were analyzed in the presence or absence of GSH depletion using L-buthionine-[S,R]-sulfoximine and iNOS induction. L-Buthionine 133-145 heme oxygenase 1 Homo sapiens 45-61 12842469-0 2003 Crystal structures of the ferric, ferrous, and ferrous-NO forms of the Asp140Ala mutant of human heme oxygenase-1: catalytic implications. Ferric enterobactin ion 26-32 heme oxygenase 1 Homo sapiens 97-113 12842469-0 2003 Crystal structures of the ferric, ferrous, and ferrous-NO forms of the Asp140Ala mutant of human heme oxygenase-1: catalytic implications. ammonium ferrous sulfate 34-41 heme oxygenase 1 Homo sapiens 97-113 12842469-0 2003 Crystal structures of the ferric, ferrous, and ferrous-NO forms of the Asp140Ala mutant of human heme oxygenase-1: catalytic implications. ferrous-no 47-57 heme oxygenase 1 Homo sapiens 97-113 12832044-3 2003 While no effect was observed in the presence of iNOS activity alone, a synergistic effect on HO-1 expression was observed in the presence of iNOS expression and GSH depletion. Glutathione 161-164 heme oxygenase 1 Homo sapiens 93-97 12839943-6 2003 In vitro studies with cultured SW480 cells, which express HO-1, showed that PEG-ZnPP induced oxidative stress, and consequently apoptotic death, of these cells. peg-znpp 76-84 heme oxygenase 1 Homo sapiens 58-62 12810075-5 2003 In addition, transfection of human Jurkat T cells with HO-1 suppressed the cellular proliferation, and this effect was reversed by zinc protoporphyrin, a specific HO competitive inhibitor. zinc protoporphyrin 131-150 heme oxygenase 1 Homo sapiens 55-59 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 14592553-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene attenuated heme and Ang II-induced prostaglandin synthesis. Heme 103-107 heme oxygenase 1 Homo sapiens 16-20 14592553-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene attenuated heme and Ang II-induced prostaglandin synthesis. Heme 103-107 heme oxygenase 1 Homo sapiens 82-86 14592553-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene attenuated heme and Ang II-induced prostaglandin synthesis. Prostaglandins 127-140 heme oxygenase 1 Homo sapiens 16-20 14592553-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene attenuated heme and Ang II-induced prostaglandin synthesis. Prostaglandins 127-140 heme oxygenase 1 Homo sapiens 82-86 12622689-0 2003 Haem and nitric oxide: synergism in the modulation of the endothelial haem oxygenase-1 pathway. Nitric Oxide 9-21 heme oxygenase 1 Homo sapiens 70-86 12622689-1 2003 NO potently up-regulates vascular haem oxygenase-1 (HO-1), an inducible defensive protein that degrades haem to CO, iron and the antioxidant bilirubin. Iron 116-120 heme oxygenase 1 Homo sapiens 34-50 12622689-1 2003 NO potently up-regulates vascular haem oxygenase-1 (HO-1), an inducible defensive protein that degrades haem to CO, iron and the antioxidant bilirubin. Iron 116-120 heme oxygenase 1 Homo sapiens 52-56 12622689-1 2003 NO potently up-regulates vascular haem oxygenase-1 (HO-1), an inducible defensive protein that degrades haem to CO, iron and the antioxidant bilirubin. Bilirubin 141-150 heme oxygenase 1 Homo sapiens 34-50 12622689-1 2003 NO potently up-regulates vascular haem oxygenase-1 (HO-1), an inducible defensive protein that degrades haem to CO, iron and the antioxidant bilirubin. Bilirubin 141-150 heme oxygenase 1 Homo sapiens 52-56 12622689-5 2003 A role for haem metabolites in modulating HO-1 expression by NO was assessed by exposing cells to SNAP, SNP or DETA/NO in medium derived from cells treated with haemin, which contained increased bilirubin levels. DEET 111-115 heme oxygenase 1 Homo sapiens 42-46 12622689-5 2003 A role for haem metabolites in modulating HO-1 expression by NO was assessed by exposing cells to SNAP, SNP or DETA/NO in medium derived from cells treated with haemin, which contained increased bilirubin levels. Hemin 161-167 heme oxygenase 1 Homo sapiens 42-46 12622689-5 2003 A role for haem metabolites in modulating HO-1 expression by NO was assessed by exposing cells to SNAP, SNP or DETA/NO in medium derived from cells treated with haemin, which contained increased bilirubin levels. Bilirubin 195-204 heme oxygenase 1 Homo sapiens 42-46 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 0-16 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 18-22 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Carbon Monoxide 78-80 heme oxygenase 1 Homo sapiens 0-16 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Carbon Monoxide 78-80 heme oxygenase 1 Homo sapiens 18-22 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Iron 83-87 heme oxygenase 1 Homo sapiens 0-16 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Iron 83-87 heme oxygenase 1 Homo sapiens 18-22 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 0-16 12704646-1 2003 Heme oxygenase-1 (HO-1) catalyzes the degradation of heme to carbon monoxide (CO), iron, and biliverdin. Biliverdine 93-103 heme oxygenase 1 Homo sapiens 18-22 12704646-3 2003 Although interest in HO-1 originally centered on its heme-degrading function, recent findings indicate that HO-1 exerts other biologically important actions. Heme 53-57 heme oxygenase 1 Homo sapiens 21-25 12704646-6 2003 Although the mechanisms responsible for the growth promoting properties of HO-1 are not well established, HO-1 can indirectly influence growth by regulating the synthesis of growth factors and by modulating the delivery of oxygen or nutrients to specific target tissues. Oxygen 223-229 heme oxygenase 1 Homo sapiens 75-79 12704646-6 2003 Although the mechanisms responsible for the growth promoting properties of HO-1 are not well established, HO-1 can indirectly influence growth by regulating the synthesis of growth factors and by modulating the delivery of oxygen or nutrients to specific target tissues. Oxygen 223-229 heme oxygenase 1 Homo sapiens 106-110 12570874-0 2003 Curcumin activates the haem oxygenase-1 gene via regulation of Nrf2 and the antioxidant-responsive element. Curcumin 0-8 heme oxygenase 1 Homo sapiens 23-39 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Heme 6-10 heme oxygenase 1 Homo sapiens 24-29 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 108-118 heme oxygenase 1 Homo sapiens 6-22 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Biliverdine 108-118 heme oxygenase 1 Homo sapiens 24-29 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 120-122 heme oxygenase 1 Homo sapiens 6-22 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Carbon Monoxide 120-122 heme oxygenase 1 Homo sapiens 24-29 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 133-137 heme oxygenase 1 Homo sapiens 6-22 12626517-1 2003 Human heme oxygenase-1 (hHO-1) catalyzes the NADPH-cytochrome P450 reductase-dependent oxidation of heme to biliverdin, CO, and free iron. Iron 133-137 heme oxygenase 1 Homo sapiens 24-29 12626517-3 2003 Earlier kinetic studies suggested that biliverdin reductase facilitates the release of biliverdin from hHO-1 (Liu, Y., and Ortiz de Montellano, P. R. (2000) J. Biol. Biliverdine 39-49 heme oxygenase 1 Homo sapiens 103-108 12626517-9 2003 Mutation of surface ionic residues shows that hHO-1 residues Lys18, Lys22, Lys179, Arg183, Arg198, Glu19, Glu127, and Glu190 contribute to the binding of cytochrome P450 reductase. octadeca(lysine) 61-66 heme oxygenase 1 Homo sapiens 46-51 12585963-4 2003 Either N-acetylcysteine, an antioxidant, or deferoxamine, an iron chelator, resulted in a dose-dependent inhibition of HO-1 mRNA and protein induction during glucose deprivation, suggesting a redox- and iron-dependent mechanism. Iron 203-207 heme oxygenase 1 Homo sapiens 119-123 12585963-5 2003 Inhibitors of electron-transport chain complex III, antimycin A and myxothiazol, the ATP synthase inhibitor oligomycin and ATP depletion with 2-deoxyglucose or glucosamine also blocked HO-1 induction. myxothiazol 68-79 heme oxygenase 1 Homo sapiens 185-189 12585963-5 2003 Inhibitors of electron-transport chain complex III, antimycin A and myxothiazol, the ATP synthase inhibitor oligomycin and ATP depletion with 2-deoxyglucose or glucosamine also blocked HO-1 induction. Oligomycins 108-118 heme oxygenase 1 Homo sapiens 185-189 12585963-5 2003 Inhibitors of electron-transport chain complex III, antimycin A and myxothiazol, the ATP synthase inhibitor oligomycin and ATP depletion with 2-deoxyglucose or glucosamine also blocked HO-1 induction. Deoxyglucose 142-156 heme oxygenase 1 Homo sapiens 185-189 12585963-5 2003 Inhibitors of electron-transport chain complex III, antimycin A and myxothiazol, the ATP synthase inhibitor oligomycin and ATP depletion with 2-deoxyglucose or glucosamine also blocked HO-1 induction. Glucosamine 160-171 heme oxygenase 1 Homo sapiens 185-189 12585963-6 2003 To address the involvement of ROS further, specifically H(2)O(2), we showed that overexpression of catalase completely blocked HO-1 activation by glucose deprivation. Reactive Oxygen Species 30-33 heme oxygenase 1 Homo sapiens 127-131 12585963-8 2003 These results demonstrate that activation of the HO-1 gene by glucose deprivation is mediated by a "glucose metabolic response" pathway via generation of ROS and that the pathway requires a functional electron-transport chain. Glucose 62-69 heme oxygenase 1 Homo sapiens 49-53 12585963-8 2003 These results demonstrate that activation of the HO-1 gene by glucose deprivation is mediated by a "glucose metabolic response" pathway via generation of ROS and that the pathway requires a functional electron-transport chain. Reactive Oxygen Species 154-157 heme oxygenase 1 Homo sapiens 49-53 12649278-0 2003 Activation of the prolyl hydroxylase oxygen-sensor results in induction of GLUT1, heme oxygenase-1, and nitric-oxide synthase proteins and confers protection from metabolic inhibition to cardiomyocytes. Oxygen 37-43 heme oxygenase 1 Homo sapiens 82-98 12649278-7 2003 In addition, ethyl-3,4 dihydroxybenzoate, dimethyloxalylglycine, and hypoxia treatments were found to induce protein levels of nitricoxide synthase-2 and heme oxygenase-1, two important cardioregulatory proteins whose expression in response to hypoxic conditions is poorly understood. ethyl protocatechuate 13-40 heme oxygenase 1 Homo sapiens 139-170 12649278-7 2003 In addition, ethyl-3,4 dihydroxybenzoate, dimethyloxalylglycine, and hypoxia treatments were found to induce protein levels of nitricoxide synthase-2 and heme oxygenase-1, two important cardioregulatory proteins whose expression in response to hypoxic conditions is poorly understood. oxalylglycine 42-63 heme oxygenase 1 Homo sapiens 139-170 12853318-10 2003 Using primary neuronal cultures, resveratrol was able to significantly induce heme oxygenase 1, whereas vehicle control showed no effect. Resveratrol 33-44 heme oxygenase 1 Homo sapiens 78-94 12570874-7 2003 From several lines of investigation we also report that curcumin (and, by inference, CAPE) stimulates ho-1 gene activity by promoting inactivation of the Nrf2-Keap1 complex, leading to increased Nrf2 binding to the resident ho-1 AREs. Curcumin 56-64 heme oxygenase 1 Homo sapiens 224-228 12570874-8 2003 Moreover, using antibodies and specific inhibitors of the mitogen-activated protein kinase (MAPK) pathways, we provide data implicating p38 MAPK in curcumin-mediated ho-1 induction. Curcumin 148-156 heme oxygenase 1 Homo sapiens 166-170 12570874-9 2003 Taken together, these results demonstrate that induction of HO-1 by curcumin and CAPE requires the activation of the Nrf2/ARE pathway. Curcumin 68-76 heme oxygenase 1 Homo sapiens 60-64 12570874-9 2003 Taken together, these results demonstrate that induction of HO-1 by curcumin and CAPE requires the activation of the Nrf2/ARE pathway. caffeic acid phenethyl ester 81-85 heme oxygenase 1 Homo sapiens 60-64 12585963-0 2003 Haem oxygenase 1 gene induction by glucose deprivation is mediated by reactive oxygen species via the mitochondrial electron-transport chain. Reactive Oxygen Species 70-93 heme oxygenase 1 Homo sapiens 0-16 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Iron 145-149 heme oxygenase 1 Homo sapiens 49-65 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Iron 145-149 heme oxygenase 1 Homo sapiens 67-71 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Carbon Monoxide 151-153 heme oxygenase 1 Homo sapiens 49-65 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Carbon Monoxide 151-153 heme oxygenase 1 Homo sapiens 67-71 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 49-65 12585963-2 2003 One such protective response is the induction of haem oxygenase 1 (HO-1), which catalyses the rate-limiting step in haem degradation, liberating iron, CO and biliverdin. Biliverdine 158-168 heme oxygenase 1 Homo sapiens 67-71 12585963-3 2003 The present study evaluated the role of ROS and the mitochondrial electron-transport chain in the induction of HO-1 by glucose deprivation in HepG2 hepatoma cells. Reactive Oxygen Species 40-43 heme oxygenase 1 Homo sapiens 111-115 12585963-4 2003 Either N-acetylcysteine, an antioxidant, or deferoxamine, an iron chelator, resulted in a dose-dependent inhibition of HO-1 mRNA and protein induction during glucose deprivation, suggesting a redox- and iron-dependent mechanism. Acetylcysteine 7-23 heme oxygenase 1 Homo sapiens 119-123 12585963-4 2003 Either N-acetylcysteine, an antioxidant, or deferoxamine, an iron chelator, resulted in a dose-dependent inhibition of HO-1 mRNA and protein induction during glucose deprivation, suggesting a redox- and iron-dependent mechanism. Deferoxamine 44-56 heme oxygenase 1 Homo sapiens 119-123 12585963-4 2003 Either N-acetylcysteine, an antioxidant, or deferoxamine, an iron chelator, resulted in a dose-dependent inhibition of HO-1 mRNA and protein induction during glucose deprivation, suggesting a redox- and iron-dependent mechanism. Iron 61-65 heme oxygenase 1 Homo sapiens 119-123 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. Curcumin 35-43 heme oxygenase 1 Homo sapiens 127-143 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. Curcumin 35-43 heme oxygenase 1 Homo sapiens 145-149 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. caffeic acid phenethyl ester 48-76 heme oxygenase 1 Homo sapiens 127-143 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. caffeic acid phenethyl ester 48-76 heme oxygenase 1 Homo sapiens 145-149 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. caffeic acid phenethyl ester 78-82 heme oxygenase 1 Homo sapiens 127-143 12570874-4 2003 Recently, the natural antioxidants curcumin and caffeic acid phenethyl ester (CAPE) have been identified as potent inducers of haem oxygenase-1 (HO-1), a redox-sensitive inducible protein that provides protection against various forms of stress. caffeic acid phenethyl ester 78-82 heme oxygenase 1 Homo sapiens 145-149 12570874-7 2003 From several lines of investigation we also report that curcumin (and, by inference, CAPE) stimulates ho-1 gene activity by promoting inactivation of the Nrf2-Keap1 complex, leading to increased Nrf2 binding to the resident ho-1 AREs. Curcumin 56-64 heme oxygenase 1 Homo sapiens 102-106 12709592-9 2003 The effect of a HO-1 enzyme activity inhibitor, tin protoporphyrin (SnPP), on Caco-2 cell proliferation and differentiation was examined. S-Nitroso-N-propionyl-D,L-penicillamine 68-72 heme oxygenase 1 Homo sapiens 16-20 12770927-12 2003 Furthermore, reducing sugars have been shown to induce oxidative stress, and ribose could be a possible cause for the upregulation of HO-1, which has been implicated in the release of angiogenic factors. Ribose 77-83 heme oxygenase 1 Homo sapiens 134-138 12709581-6 2003 Elevation of HO-1 protein was accompanied by a marked increase in catalytic activity of the enzyme as reflected by enhanced formation of both carbon monoxide and the endogenous antioxidant, bilirubin. Carbon Monoxide 142-157 heme oxygenase 1 Homo sapiens 13-17 12709581-6 2003 Elevation of HO-1 protein was accompanied by a marked increase in catalytic activity of the enzyme as reflected by enhanced formation of both carbon monoxide and the endogenous antioxidant, bilirubin. Bilirubin 190-199 heme oxygenase 1 Homo sapiens 13-17 12800860-1 2003 Haem-oxygenase-1 (HO-1) is an antioxidant stress protein that is mainly induced by reactive oxygen species, inflammatory cytokines and hyperthermia. Reactive Oxygen Species 83-106 heme oxygenase 1 Homo sapiens 0-16 12709581-10 2003 Increased HO-1 expression and ensuing formation of bilirubin and carbon monoxide may contribute to and explain the specific antioxidant and antiatherogenic actions of PETN. Carbon Monoxide 65-80 heme oxygenase 1 Homo sapiens 10-14 12709582-0 2003 Influence of heme and heme oxygenase-1 transfection of pulmonary microvascular endothelium on oxidant generation and cGMP. Cyclic GMP 117-121 heme oxygenase 1 Homo sapiens 22-38 12709582-3 2003 Culture of PMEC with low serum heme decreased cGMP and the detection of peroxide with 10 microM 2",7"-dichlorofluorescin diacetate and increased HO-1 further decreased cGMP without altering the peroxide detection under these conditions. Heme 31-35 heme oxygenase 1 Homo sapiens 145-149 12709582-3 2003 Culture of PMEC with low serum heme decreased cGMP and the detection of peroxide with 10 microM 2",7"-dichlorofluorescin diacetate and increased HO-1 further decreased cGMP without altering the peroxide detection under these conditions. Cyclic GMP 168-172 heme oxygenase 1 Homo sapiens 145-149 12709585-2 2003 Although most of the monocyte-derived cytokines exhibit proinflammatory functions in vivo, heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, exerts potent anti-inflammatory effect through production of carbon monoxide and bilirubin. Heme 91-95 heme oxygenase 1 Homo sapiens 109-113 12709585-2 2003 Although most of the monocyte-derived cytokines exhibit proinflammatory functions in vivo, heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, exerts potent anti-inflammatory effect through production of carbon monoxide and bilirubin. Carbon Monoxide 213-228 heme oxygenase 1 Homo sapiens 91-107 12709585-2 2003 Although most of the monocyte-derived cytokines exhibit proinflammatory functions in vivo, heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, exerts potent anti-inflammatory effect through production of carbon monoxide and bilirubin. Carbon Monoxide 213-228 heme oxygenase 1 Homo sapiens 109-113 12709585-2 2003 Although most of the monocyte-derived cytokines exhibit proinflammatory functions in vivo, heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, exerts potent anti-inflammatory effect through production of carbon monoxide and bilirubin. Bilirubin 233-242 heme oxygenase 1 Homo sapiens 91-107 12709585-2 2003 Although most of the monocyte-derived cytokines exhibit proinflammatory functions in vivo, heme oxygenase-1 (HO-1), an inducible heme-degrading enzyme, exerts potent anti-inflammatory effect through production of carbon monoxide and bilirubin. Bilirubin 233-242 heme oxygenase 1 Homo sapiens 109-113 12709592-8 2003 HO-1 activity was determined by the ability of the enzyme to generate bilirubin from hemin. Bilirubin 70-79 heme oxygenase 1 Homo sapiens 0-4 12709592-8 2003 HO-1 activity was determined by the ability of the enzyme to generate bilirubin from hemin. Hemin 85-90 heme oxygenase 1 Homo sapiens 0-4 12709592-9 2003 The effect of a HO-1 enzyme activity inhibitor, tin protoporphyrin (SnPP), on Caco-2 cell proliferation and differentiation was examined. tin protoporphyrin IX 48-66 heme oxygenase 1 Homo sapiens 16-20 12720199-1 2003 BACKGROUND/PURPOSE: Heme oxygenase (HO-1), an inducible isoform of HO is a regulator of vascular tone and cell proliferation through the production of endogenous carbon monoxide (CO). Carbon Monoxide 162-177 heme oxygenase 1 Homo sapiens 36-40 12720199-1 2003 BACKGROUND/PURPOSE: Heme oxygenase (HO-1), an inducible isoform of HO is a regulator of vascular tone and cell proliferation through the production of endogenous carbon monoxide (CO). Carbon Monoxide 179-181 heme oxygenase 1 Homo sapiens 36-40 12720199-10 2003 HO-1 and eNOS immunoreactivity was reduced markedly reduced in the endothelium and arterial wall in the CDH samples compared with normal lung. CDw17 antigen 104-107 heme oxygenase 1 Homo sapiens 0-4 12709568-2 2003 We examined the effect of delivery of the human HO-1 gene on cellular heme in renal tissue using a retroviral vector. Heme 70-74 heme oxygenase 1 Homo sapiens 48-52 12709568-9 2003 Compared with nontransduced rats, human HO-1 gene overexpression in transduced rats was associated with a 35% decrease in urinary 20-hydroxyeicosatetraenoic acid, a potent vasoconstrictor and an inhibitor of tubular Na(+) transport, which may be related to the decrease in blood pressure. 20-hydroxy-5,8,11,14-eicosatetraenoic acid 130-161 heme oxygenase 1 Homo sapiens 40-44 12709570-5 2003 This plasmid contains the H-2K(b) promoter, which transcribes the tet transactivator (tTA) and expression of a human HO-1 cDNA is obtained in the absence of tetracycline. Tetracycline 157-169 heme oxygenase 1 Homo sapiens 117-121 12709571-1 2003 Heme oxygenase-1 (HO-1) is an essential enzyme in heme catabolism and is characterized by its inducibility in response to various environmental factors, including its substrate heme. Heme 50-54 heme oxygenase 1 Homo sapiens 0-16 12709571-1 2003 Heme oxygenase-1 (HO-1) is an essential enzyme in heme catabolism and is characterized by its inducibility in response to various environmental factors, including its substrate heme. Heme 50-54 heme oxygenase 1 Homo sapiens 18-22 12709571-4 2003 The downregulation of HO-1 expression may reduce energy expenditure and local production of carbon monoxide, iron, and bilirubin and transiently increase intracellular heme pool. Carbon Monoxide 92-107 heme oxygenase 1 Homo sapiens 22-26 12709571-4 2003 The downregulation of HO-1 expression may reduce energy expenditure and local production of carbon monoxide, iron, and bilirubin and transiently increase intracellular heme pool. Iron 109-113 heme oxygenase 1 Homo sapiens 22-26 12709571-4 2003 The downregulation of HO-1 expression may reduce energy expenditure and local production of carbon monoxide, iron, and bilirubin and transiently increase intracellular heme pool. Bilirubin 119-128 heme oxygenase 1 Homo sapiens 22-26 12709571-4 2003 The downregulation of HO-1 expression may reduce energy expenditure and local production of carbon monoxide, iron, and bilirubin and transiently increase intracellular heme pool. Heme 168-172 heme oxygenase 1 Homo sapiens 22-26 12800860-1 2003 Haem-oxygenase-1 (HO-1) is an antioxidant stress protein that is mainly induced by reactive oxygen species, inflammatory cytokines and hyperthermia. Reactive Oxygen Species 83-106 heme oxygenase 1 Homo sapiens 18-22 12678694-6 2003 HO1-mediated metabolism of heme groups released from NO-damaged proteins leads to a change in the levels of redox-active iron and a release of carbon monoxide (CO) and bilirubin, all of which have been implicated in cellular resistance to oxidative stress. Heme 27-31 heme oxygenase 1 Homo sapiens 0-3 12578834-0 2003 Nerve growth factor protects against 6-hydroxydopamine-induced oxidative stress by increasing expression of heme oxygenase-1 in a phosphatidylinositol 3-kinase-dependent manner. Oxidopamine 37-54 heme oxygenase 1 Homo sapiens 108-124 12578834-4 2003 The effect of NGF was mimicked by induction of HO-1 expression with CoCl(2); by treatment with bilirubin, an end product of heme catabolism; and by infection with a retroviral expression vector for human HO-1. cocl( 68-73 heme oxygenase 1 Homo sapiens 47-51 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Reactive Oxygen Species 37-40 heme oxygenase 1 Homo sapiens 17-21 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Reactive Oxygen Species 37-40 heme oxygenase 1 Homo sapiens 120-124 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. tin protoporphyrin IX 135-153 heme oxygenase 1 Homo sapiens 17-21 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. tin protoporphyrin IX 135-153 heme oxygenase 1 Homo sapiens 120-124 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Reactive Oxygen Species 240-243 heme oxygenase 1 Homo sapiens 17-21 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Reactive Oxygen Species 240-243 heme oxygenase 1 Homo sapiens 120-124 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Oxidopamine 267-273 heme oxygenase 1 Homo sapiens 17-21 12578834-5 2003 The relevance of HO-1 in NGF-induced ROS reduction was further demonstrated by the evidence that cells treated with the HO-1 inhibitor tin-protoporphyrin or infected with a retroviral expression vector for antisense HO-1 exhibited enhanced ROS release in response to 6-OHDA, despite the presence of the neurotrophin. Oxidopamine 267-273 heme oxygenase 1 Homo sapiens 120-124 12578834-8 2003 These observations indicate that the PI3K/Akt pathway controls the intracellular levels of ROS by regulating the expression of the antioxidant enzyme HO-1. Reactive Oxygen Species 91-94 heme oxygenase 1 Homo sapiens 150-154 12727804-7 2003 Genes whose transcription was altered in common among all the treated keratinocytes included those induced by reactive oxygen, of which heme oxygenase-1 displayed the highest fold induction. reactive oxygen 110-125 heme oxygenase 1 Homo sapiens 136-152 12727804-10 2003 Comparison with other agents producing reactive oxygen in the cells, as reflected in heme oxygenase-1 induction, suggested cellular differentiation was suppressed by sustained but not transient generation of reactive oxygen. reactive 39-47 heme oxygenase 1 Homo sapiens 85-101 12727804-10 2003 Comparison with other agents producing reactive oxygen in the cells, as reflected in heme oxygenase-1 induction, suggested cellular differentiation was suppressed by sustained but not transient generation of reactive oxygen. Oxygen 48-54 heme oxygenase 1 Homo sapiens 85-101 12678694-6 2003 HO1-mediated metabolism of heme groups released from NO-damaged proteins leads to a change in the levels of redox-active iron and a release of carbon monoxide (CO) and bilirubin, all of which have been implicated in cellular resistance to oxidative stress. Iron 121-125 heme oxygenase 1 Homo sapiens 0-3 12678694-6 2003 HO1-mediated metabolism of heme groups released from NO-damaged proteins leads to a change in the levels of redox-active iron and a release of carbon monoxide (CO) and bilirubin, all of which have been implicated in cellular resistance to oxidative stress. Carbon Monoxide 143-158 heme oxygenase 1 Homo sapiens 0-3 12678694-6 2003 HO1-mediated metabolism of heme groups released from NO-damaged proteins leads to a change in the levels of redox-active iron and a release of carbon monoxide (CO) and bilirubin, all of which have been implicated in cellular resistance to oxidative stress. Carbon Monoxide 160-162 heme oxygenase 1 Homo sapiens 0-3 12678694-6 2003 HO1-mediated metabolism of heme groups released from NO-damaged proteins leads to a change in the levels of redox-active iron and a release of carbon monoxide (CO) and bilirubin, all of which have been implicated in cellular resistance to oxidative stress. Bilirubin 168-177 heme oxygenase 1 Homo sapiens 0-3 12676598-5 2003 HO-1 expression, a sensitive marker for oxidative stress, is directly correlated with the high organic carbon and polycyclic aromatic hydrocarbon (PAH) content of UFPs. Carbon 103-109 heme oxygenase 1 Homo sapiens 0-4 12678694-7 2003 Perhaps one or more of the products of HO1 heme metabolism is involved in induced adaptive resistance or perhaps a heme-independent mechanism is involved. Heme 43-47 heme oxygenase 1 Homo sapiens 39-42 12676598-5 2003 HO-1 expression, a sensitive marker for oxidative stress, is directly correlated with the high organic carbon and polycyclic aromatic hydrocarbon (PAH) content of UFPs. Polycyclic Aromatic Hydrocarbons 114-145 heme oxygenase 1 Homo sapiens 0-4 12676598-5 2003 HO-1 expression, a sensitive marker for oxidative stress, is directly correlated with the high organic carbon and polycyclic aromatic hydrocarbon (PAH) content of UFPs. Polycyclic Aromatic Hydrocarbons 147-150 heme oxygenase 1 Homo sapiens 0-4 12572666-6 2003 Heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in Alzheimer disease suggesting increased heme turnover as a source of redox-active iron. Biliverdine 78-88 heme oxygenase 1 Homo sapiens 0-16 12676598-6 2003 The dithiothreitol (DTT) assay, a quantitative measure of in vitro ROS formation, was correlated with PAH content and HO-1 expression. Dithiothreitol 4-18 heme oxygenase 1 Homo sapiens 118-122 12676598-6 2003 The dithiothreitol (DTT) assay, a quantitative measure of in vitro ROS formation, was correlated with PAH content and HO-1 expression. Dithiothreitol 20-23 heme oxygenase 1 Homo sapiens 118-122 12676598-6 2003 The dithiothreitol (DTT) assay, a quantitative measure of in vitro ROS formation, was correlated with PAH content and HO-1 expression. Reactive Oxygen Species 67-70 heme oxygenase 1 Homo sapiens 118-122 12500973-0 2003 Comparison of the heme-free and -bound crystal structures of human heme oxygenase-1. Heme 18-22 heme oxygenase 1 Homo sapiens 67-83 12594185-9 2003 HO-1 plays an important protective role in the defense against reactive oxygen species (ROS). Reactive Oxygen Species 63-86 heme oxygenase 1 Homo sapiens 0-4 12594185-9 2003 HO-1 plays an important protective role in the defense against reactive oxygen species (ROS). Reactive Oxygen Species 88-91 heme oxygenase 1 Homo sapiens 0-4 12668974-0 2003 Nitric oxide-mediated cytoprotection of hepatocytes from glucose deprivation-induced cytotoxicity: involvement of heme oxygenase-1. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 114-130 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 105-120 heme oxygenase 1 Homo sapiens 0-16 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 105-120 heme oxygenase 1 Homo sapiens 18-22 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 122-124 heme oxygenase 1 Homo sapiens 0-16 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Carbon Monoxide 122-124 heme oxygenase 1 Homo sapiens 18-22 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 0-16 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 18-22 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Iron 148-152 heme oxygenase 1 Homo sapiens 0-16 12668974-1 2003 Heme oxygenase-1 (HO-1) is the rate-limiting enzyme in heme catabolism, which leads to the generation of carbon monoxide (CO), biliverdin, and free iron. Iron 148-152 heme oxygenase 1 Homo sapiens 18-22 12511571-1 2003 Heme oxygenase 1 (HO-1) catalyzes heme breakdown, eventually releasing iron, carbon monoxide, and bilirubin IXalpha. Iron 71-75 heme oxygenase 1 Homo sapiens 0-16 12511571-1 2003 Heme oxygenase 1 (HO-1) catalyzes heme breakdown, eventually releasing iron, carbon monoxide, and bilirubin IXalpha. Carbon Monoxide 77-92 heme oxygenase 1 Homo sapiens 0-16 12511571-1 2003 Heme oxygenase 1 (HO-1) catalyzes heme breakdown, eventually releasing iron, carbon monoxide, and bilirubin IXalpha. Bilirubin 98-107 heme oxygenase 1 Homo sapiens 0-16 12511571-2 2003 HO-1 is induced by its substrate heme and various environmental factors, which represents a protective response against oxidative stresses. Heme 33-37 heme oxygenase 1 Homo sapiens 0-4 12511571-6 2003 Expression of HO-1 was also reduced in human cells when exposed to interferon-gamma or an iron chelator desferrioxamine, each of which induced Bach1 expression. Iron 90-94 heme oxygenase 1 Homo sapiens 14-18 12511571-6 2003 Expression of HO-1 was also reduced in human cells when exposed to interferon-gamma or an iron chelator desferrioxamine, each of which induced Bach1 expression. Deferoxamine 104-119 heme oxygenase 1 Homo sapiens 14-18 12511571-7 2003 In contrast, induction of HO-1 expression by CoCl(2) is associated with reduced expression of Bach1 mRNA. cobaltous chloride 45-52 heme oxygenase 1 Homo sapiens 26-30 12511571-10 2003 Therefore, Bach1 functions as a hypoxia-inducible repressor for the HO-1 gene, thereby contributing to fine-tuning of oxygen homeostasis in human cells. Oxygen 118-124 heme oxygenase 1 Homo sapiens 68-72 12572666-6 2003 Heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in Alzheimer disease suggesting increased heme turnover as a source of redox-active iron. Heme 61-65 heme oxygenase 1 Homo sapiens 0-16 12572666-6 2003 Heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in Alzheimer disease suggesting increased heme turnover as a source of redox-active iron. Iron 69-73 heme oxygenase 1 Homo sapiens 0-16 12572666-6 2003 Heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in Alzheimer disease suggesting increased heme turnover as a source of redox-active iron. Heme 145-149 heme oxygenase 1 Homo sapiens 0-16 12572666-6 2003 Heme oxygenase-1, an enzyme that catalyzes the conversion of heme to iron and biliverdin, is increased in Alzheimer disease suggesting increased heme turnover as a source of redox-active iron. Iron 187-191 heme oxygenase 1 Homo sapiens 0-16 12586756-10 2003 Endogenously produced cGMP also induced HO-1 because phosphodiesterase inhibition markedly elevated HO-1. 8-bromocyclic GMP 22-26 heme oxygenase 1 Homo sapiens 40-44 12586756-10 2003 Endogenously produced cGMP also induced HO-1 because phosphodiesterase inhibition markedly elevated HO-1. 8-bromocyclic GMP 22-26 heme oxygenase 1 Homo sapiens 100-104 12586756-14 2003 Abrogation of HO-1 induction by PD-98059 showed also a role for ERK. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 32-40 heme oxygenase 1 Homo sapiens 14-18 12531245-8 2003 Endotoxin and SnMP showed more prominent effect on the expression of VEGF and suppression of TGF-beta in HO-1 transduced endothelial cells, suggesting that their effect is most probably mediated through induction of HO-1. tin mesoporphyrin 14-18 heme oxygenase 1 Homo sapiens 105-109 12623985-3 2003 In the present study, we determined the effect of interventions that suppress HO activity or induce HO-1 gene expression on Ang II-mediated increase in 6-keto-PGF1alpha and PGE2 in cultures of human femoral artery endothelial cells. 6-Ketoprostaglandin F1 alpha 152-168 heme oxygenase 1 Homo sapiens 100-104 12623985-3 2003 In the present study, we determined the effect of interventions that suppress HO activity or induce HO-1 gene expression on Ang II-mediated increase in 6-keto-PGF1alpha and PGE2 in cultures of human femoral artery endothelial cells. Dinoprostone 173-177 heme oxygenase 1 Homo sapiens 100-104 12623985-5 2003 This effect of Ang II on prostaglandin production by endothelial cells was attenuated in cells treated with SnCl2 (10 micromol/L), an inducer of HO-1, but was magnified in cells treated with the HO inhibitor ZnDPP or heme. Prostaglandins 25-38 heme oxygenase 1 Homo sapiens 145-149 12623985-5 2003 This effect of Ang II on prostaglandin production by endothelial cells was attenuated in cells treated with SnCl2 (10 micromol/L), an inducer of HO-1, but was magnified in cells treated with the HO inhibitor ZnDPP or heme. stannous chloride 108-113 heme oxygenase 1 Homo sapiens 145-149 12531245-8 2003 Endotoxin and SnMP showed more prominent effect on the expression of VEGF and suppression of TGF-beta in HO-1 transduced endothelial cells, suggesting that their effect is most probably mediated through induction of HO-1. tin mesoporphyrin 14-18 heme oxygenase 1 Homo sapiens 216-220 12623985-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene also attenuated heme and Ang II-induced prostaglandin synthesis. Heme 108-112 heme oxygenase 1 Homo sapiens 16-20 12623985-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene also attenuated heme and Ang II-induced prostaglandin synthesis. Heme 108-112 heme oxygenase 1 Homo sapiens 82-86 12591764-0 2003 Rapamycin induces heme oxygenase-1 in human pulmonary vascular cells: implications in the antiproliferative response to rapamycin. Sirolimus 0-9 heme oxygenase 1 Homo sapiens 18-34 12623985-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene also attenuated heme and Ang II-induced prostaglandin synthesis. Prostaglandins 132-145 heme oxygenase 1 Homo sapiens 16-20 12623985-6 2003 Upregulation of HO-1 gene expression by retrovirus-mediated delivery of the human HO-1 gene also attenuated heme and Ang II-induced prostaglandin synthesis. Prostaglandins 132-145 heme oxygenase 1 Homo sapiens 82-86 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Heme 76-80 heme oxygenase 1 Homo sapiens 0-16 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Heme 76-80 heme oxygenase 1 Homo sapiens 18-22 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Biliverdine 84-94 heme oxygenase 1 Homo sapiens 0-16 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Biliverdine 84-94 heme oxygenase 1 Homo sapiens 18-22 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Iron 101-105 heme oxygenase 1 Homo sapiens 0-16 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Iron 101-105 heme oxygenase 1 Homo sapiens 18-22 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 0-16 12935634-1 2003 Heme oxygenase-1 (HO-1) is a 32 kDa heat shock protein (HSP) that catalyzes heme to biliverdin, free iron and carbon monoxide in the brain. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 18-22 12591764-0 2003 Rapamycin induces heme oxygenase-1 in human pulmonary vascular cells: implications in the antiproliferative response to rapamycin. Sirolimus 120-129 heme oxygenase 1 Homo sapiens 18-34 12591764-3 2003 In this study, we evaluated whether there is an interaction between rapamycin and HO-1. Sirolimus 68-77 heme oxygenase 1 Homo sapiens 82-86 12591764-4 2003 METHODS AND RESULTS: In human pulmonary artery endothelial or smooth muscle cells, HO-1 expression was evaluated in response to rapamycin or wortmannin, an inhibitor of the upstream modulator of mammalian target of rapamycin (mTOR) PI-3K. Sirolimus 128-137 heme oxygenase 1 Homo sapiens 83-87 12591764-4 2003 METHODS AND RESULTS: In human pulmonary artery endothelial or smooth muscle cells, HO-1 expression was evaluated in response to rapamycin or wortmannin, an inhibitor of the upstream modulator of mammalian target of rapamycin (mTOR) PI-3K. Wortmannin 141-151 heme oxygenase 1 Homo sapiens 83-87 12591764-6 2003 Rapamycin induced HO-1 expression in both pulmonary endothelial and smooth muscle cells, whereas no to little increase was seen in response to another immunosuppressive agent, cyclosporin A. Sirolimus 0-9 heme oxygenase 1 Homo sapiens 18-22 12591764-7 2003 HO-1 expression was also increased in response to wortmannin, suggesting that the PI-3K-mTOR pathway is required for this induction. Wortmannin 50-60 heme oxygenase 1 Homo sapiens 0-4 12591764-9 2003 CONCLUSIONS: The induction of HO-1 expression by rapamycin and, more importantly, the effects of tin protoporphyrin, an inhibitor of HO activity, on the antiproliferative actions of rapamycin suggest that the effects of rapamycin may be, at least in part, modulated by its actions on HO-1. tin protoporphyrin IX 97-115 heme oxygenase 1 Homo sapiens 284-288 12591764-9 2003 CONCLUSIONS: The induction of HO-1 expression by rapamycin and, more importantly, the effects of tin protoporphyrin, an inhibitor of HO activity, on the antiproliferative actions of rapamycin suggest that the effects of rapamycin may be, at least in part, modulated by its actions on HO-1. Sirolimus 49-58 heme oxygenase 1 Homo sapiens 30-34 12591764-9 2003 CONCLUSIONS: The induction of HO-1 expression by rapamycin and, more importantly, the effects of tin protoporphyrin, an inhibitor of HO activity, on the antiproliferative actions of rapamycin suggest that the effects of rapamycin may be, at least in part, modulated by its actions on HO-1. Sirolimus 182-191 heme oxygenase 1 Homo sapiens 30-34 12591764-9 2003 CONCLUSIONS: The induction of HO-1 expression by rapamycin and, more importantly, the effects of tin protoporphyrin, an inhibitor of HO activity, on the antiproliferative actions of rapamycin suggest that the effects of rapamycin may be, at least in part, modulated by its actions on HO-1. Sirolimus 182-191 heme oxygenase 1 Homo sapiens 284-288 12420309-4 2002 Catecholamines caused a reduction in intracellular glutathione levels, an accumulation in reactive oxygen species and in heme oxygenase-1, the 32 kDa stress-induced protein. Catecholamines 0-14 heme oxygenase 1 Homo sapiens 121-137 12566071-0 2003 Beta-carotene suppresses UVA-induced HO-1 gene expression in cultured FEK4. beta Carotene 0-13 heme oxygenase 1 Homo sapiens 37-41 12566071-4 2003 Carotenoids, as the most potent singlet oxygen quenchers in nature, are expected to effectively suppress the UVA-induced HO-1 gene activation in human cells. Carotenoids 0-11 heme oxygenase 1 Homo sapiens 121-125 12566071-5 2003 In this study, we measured the suppression of UVA-induced levels of HO-1 mRNA after the addition of a series of six all-trans-beta-carotene concentrations (0.07, 0.2, 0.8, 2.3, 8.0, and 21 microM) to the culture medium of exponentially growing FEK4 cells. beta Carotene 126-139 heme oxygenase 1 Homo sapiens 68-72 12566071-7 2003 The results of this study show a concentration-dependent suppression of UVA- (250 kJ/m(2)) induced transcriptional activation of HO-1 in exponentially growing FEK4 cells by beta-carotene. beta Carotene 173-186 heme oxygenase 1 Homo sapiens 129-133 21207851-1 2003 AIM: To explore the effects of heme- heme oxygenase-1 (HO-1)-carbon monoxide(CO)-cyclic GMP (cGMP)on aortic vascular reactivity in endotoxemic rats and its molecular mechanism. Carbon Monoxide 61-76 heme oxygenase 1 Homo sapiens 55-59 21207851-1 2003 AIM: To explore the effects of heme- heme oxygenase-1 (HO-1)-carbon monoxide(CO)-cyclic GMP (cGMP)on aortic vascular reactivity in endotoxemic rats and its molecular mechanism. Carbon Monoxide 77-80 heme oxygenase 1 Homo sapiens 55-59 21207851-1 2003 AIM: To explore the effects of heme- heme oxygenase-1 (HO-1)-carbon monoxide(CO)-cyclic GMP (cGMP)on aortic vascular reactivity in endotoxemic rats and its molecular mechanism. Cyclic GMP 81-91 heme oxygenase 1 Homo sapiens 55-59 21207851-1 2003 AIM: To explore the effects of heme- heme oxygenase-1 (HO-1)-carbon monoxide(CO)-cyclic GMP (cGMP)on aortic vascular reactivity in endotoxemic rats and its molecular mechanism. Cyclic GMP 93-97 heme oxygenase 1 Homo sapiens 55-59 14753445-0 2003 Transient elevation of serum bilirubin (a heme oxygenase-1 metabolite) level in hemorrhagic stroke: bilirubin is a marker of oxidant stress. Bilirubin 29-38 heme oxygenase 1 Homo sapiens 42-58 14753445-0 2003 Transient elevation of serum bilirubin (a heme oxygenase-1 metabolite) level in hemorrhagic stroke: bilirubin is a marker of oxidant stress. Bilirubin 100-109 heme oxygenase 1 Homo sapiens 42-58 14753445-2 2003 The production of Bil reflects heme oxygenase-1 expression in response to oxidative stress in various diseases. Bilirubin 18-21 heme oxygenase 1 Homo sapiens 31-47 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Oxygen 33-39 heme oxygenase 1 Homo sapiens 0-21 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Oxygen 33-39 heme oxygenase 1 Homo sapiens 23-28 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. C-1027 AROMATIZED CHROMOPHORE 89-92 heme oxygenase 1 Homo sapiens 0-21 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. C-1027 AROMATIZED CHROMOPHORE 89-92 heme oxygenase 1 Homo sapiens 23-28 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Heme 107-111 heme oxygenase 1 Homo sapiens 0-21 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Heme 107-111 heme oxygenase 1 Homo sapiens 23-28 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Bilirubin 152-161 heme oxygenase 1 Homo sapiens 0-21 15115285-1 2003 Heat shock protein 32 (Hsp32, hemoxygenase-1) is induced by reactive oxygen metabolites (ROM) and degrades heme leading to the formation of antioxidant bilirubin. Bilirubin 152-161 heme oxygenase 1 Homo sapiens 23-28 15115285-6 2003 Hsp32 expression in formalin-fixed sections was assessed by avidin-biotin peroxidase immunohistochemistry using a polyclonal rabbit anti-Hsp32 as the primary antibody. Formaldehyde 20-28 heme oxygenase 1 Homo sapiens 0-5 12551835-0 2003 Contribution of the induction of heme oxygenase-1 to etoposide-induced apoptosis in acute myeloblastic leukemia. Etoposide 53-62 heme oxygenase 1 Homo sapiens 33-49 12964953-2 2003 In addition to its physiological role in heme degradation, HO-1 may influence a number of cellular processes, including growth, inflammation, and apoptosis. Heme 41-45 heme oxygenase 1 Homo sapiens 59-63 12964953-4 2003 The transcriptional upregulation of HO-1 responds to many agents, such as hypoxia, bacterial lipopolysaccharide, and reactive oxygen/nitrogen species. bacterial lipopolysaccharide 83-111 heme oxygenase 1 Homo sapiens 36-40 12964953-4 2003 The transcriptional upregulation of HO-1 responds to many agents, such as hypoxia, bacterial lipopolysaccharide, and reactive oxygen/nitrogen species. reactive oxygen/nitrogen species 117-149 heme oxygenase 1 Homo sapiens 36-40 12964953-5 2003 HO-1 and its constitutively expressed isozyme, heme oxygenase-2, catalyze the rate-limiting step in the conversion of heme to its metabolites, bilirubin IXalpha, ferrous iron, and carbon monoxide (CO). Heme 47-51 heme oxygenase 1 Homo sapiens 0-4 12964953-5 2003 HO-1 and its constitutively expressed isozyme, heme oxygenase-2, catalyze the rate-limiting step in the conversion of heme to its metabolites, bilirubin IXalpha, ferrous iron, and carbon monoxide (CO). Bilirubin 143-160 heme oxygenase 1 Homo sapiens 0-4 12964953-5 2003 HO-1 and its constitutively expressed isozyme, heme oxygenase-2, catalyze the rate-limiting step in the conversion of heme to its metabolites, bilirubin IXalpha, ferrous iron, and carbon monoxide (CO). Iron 170-174 heme oxygenase 1 Homo sapiens 0-4 12964953-5 2003 HO-1 and its constitutively expressed isozyme, heme oxygenase-2, catalyze the rate-limiting step in the conversion of heme to its metabolites, bilirubin IXalpha, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 180-195 heme oxygenase 1 Homo sapiens 0-4 12964953-5 2003 HO-1 and its constitutively expressed isozyme, heme oxygenase-2, catalyze the rate-limiting step in the conversion of heme to its metabolites, bilirubin IXalpha, ferrous iron, and carbon monoxide (CO). Carbon Monoxide 197-199 heme oxygenase 1 Homo sapiens 0-4 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 33-37 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Carbon Monoxide 120-135 heme oxygenase 1 Homo sapiens 193-197 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Carbon Monoxide 137-139 heme oxygenase 1 Homo sapiens 33-37 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Carbon Monoxide 137-139 heme oxygenase 1 Homo sapiens 193-197 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Heme 174-178 heme oxygenase 1 Homo sapiens 33-37 12444034-2 2002 Although the mechanisms by which HO-1 exerts its cytoprotection are not clearly understood, it has been speculated that carbon monoxide (CO), a catalytic byproduct following heme catabolism by HO-1, may mediate cellular cytoprotection via its anti-inflammatory properties. Heme 174-178 heme oxygenase 1 Homo sapiens 193-197 12699247-0 2002 Interaction of heme with nitroxyl or nitric oxide amplifies heme oxygenase-1 induction: involvement of the transcription factor Nrf2. Heme 15-19 heme oxygenase 1 Homo sapiens 60-76 12699247-0 2002 Interaction of heme with nitroxyl or nitric oxide amplifies heme oxygenase-1 induction: involvement of the transcription factor Nrf2. nitroxyl 25-33 heme oxygenase 1 Homo sapiens 60-76 12699247-0 2002 Interaction of heme with nitroxyl or nitric oxide amplifies heme oxygenase-1 induction: involvement of the transcription factor Nrf2. Nitric Oxide 37-49 heme oxygenase 1 Homo sapiens 60-76 12699247-1 2002 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme, the expression of which is highly sensitive to induction by pro-oxidant stimuli including the substrate heme and reactive oxygen species. Heme 156-160 heme oxygenase 1 Homo sapiens 0-16 12699247-1 2002 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme, the expression of which is highly sensitive to induction by pro-oxidant stimuli including the substrate heme and reactive oxygen species. Heme 156-160 heme oxygenase 1 Homo sapiens 18-22 12699247-1 2002 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme, the expression of which is highly sensitive to induction by pro-oxidant stimuli including the substrate heme and reactive oxygen species. Reactive Oxygen Species 165-188 heme oxygenase 1 Homo sapiens 0-16 12699247-1 2002 Heme oxygenase-1 (HO-1) is a cytoprotective enzyme, the expression of which is highly sensitive to induction by pro-oxidant stimuli including the substrate heme and reactive oxygen species. Reactive Oxygen Species 165-188 heme oxygenase 1 Homo sapiens 18-22 12699247-2 2002 Conceptually, the perception that HO-1 plays a key role in response to oxidative damage is paralleled by evidence showing high expression of HO-1 in a variety of cell systems challenged with nitric oxide (NO) or NO-derivatives, thus revealing a potential biological function for HO-1 against nitrosative stress. Nitric Oxide 191-203 heme oxygenase 1 Homo sapiens 34-38 12699247-2 2002 Conceptually, the perception that HO-1 plays a key role in response to oxidative damage is paralleled by evidence showing high expression of HO-1 in a variety of cell systems challenged with nitric oxide (NO) or NO-derivatives, thus revealing a potential biological function for HO-1 against nitrosative stress. Nitric Oxide 191-203 heme oxygenase 1 Homo sapiens 141-145 12699247-2 2002 Conceptually, the perception that HO-1 plays a key role in response to oxidative damage is paralleled by evidence showing high expression of HO-1 in a variety of cell systems challenged with nitric oxide (NO) or NO-derivatives, thus revealing a potential biological function for HO-1 against nitrosative stress. Nitric Oxide 191-203 heme oxygenase 1 Homo sapiens 141-145 12699247-3 2002 In this study, we report that exposure of cardiac cells to hemin (5-20 microM) in combination with compounds that liberate nitroxyl (HNO/NO-) or release NO significantly potentiates HO-1 mRNA and protein expression leading to a remarkable increase in heme oxygenase activity under both normoxic and hypoxic conditions. Hemin 59-64 heme oxygenase 1 Homo sapiens 182-186 12699247-3 2002 In this study, we report that exposure of cardiac cells to hemin (5-20 microM) in combination with compounds that liberate nitroxyl (HNO/NO-) or release NO significantly potentiates HO-1 mRNA and protein expression leading to a remarkable increase in heme oxygenase activity under both normoxic and hypoxic conditions. nitroxyl 123-131 heme oxygenase 1 Homo sapiens 182-186 12699247-3 2002 In this study, we report that exposure of cardiac cells to hemin (5-20 microM) in combination with compounds that liberate nitroxyl (HNO/NO-) or release NO significantly potentiates HO-1 mRNA and protein expression leading to a remarkable increase in heme oxygenase activity under both normoxic and hypoxic conditions. nitroxyl 133-136 heme oxygenase 1 Homo sapiens 182-186 12699247-4 2002 The amplification of the heme oxygenase pathway appears to involve a direct interaction between heme and the NO groups, as the ability of both NO(-)- and NO-releasing agents to induce HO-1 is totally lost by their pre-incubation for 1 hr in complete medium prior to cell treatment but is highly preserved by addition of hemin during the preincubation step. Heme 25-29 heme oxygenase 1 Homo sapiens 184-188 12699247-4 2002 The amplification of the heme oxygenase pathway appears to involve a direct interaction between heme and the NO groups, as the ability of both NO(-)- and NO-releasing agents to induce HO-1 is totally lost by their pre-incubation for 1 hr in complete medium prior to cell treatment but is highly preserved by addition of hemin during the preincubation step. Hemin 320-325 heme oxygenase 1 Homo sapiens 184-188 12699247-8 2002 We propose that modification of the iron protoporphyrin centers by NO groups to modulate HO-1 expression might be regarded as a molecular switch to maximize heme oxygenase enzymatic activity and consequently mitigate the redox imbalance imposed by oxidative and nitrosative stress. Heme 36-55 heme oxygenase 1 Homo sapiens 89-93 12646399-6 2003 Pretreatment of monolayers with hemin for 2 h followed by 18 h in complete medium resulted in HO-1 induction and the attenuation of H(2)O(2)-mediated increases in endothelial permeability, and significantly improved the restoration of endothelial barrier function 48 h later. Hemin 32-37 heme oxygenase 1 Homo sapiens 94-98 12646399-8 2003 This potentiation was inhibited by incubation with the HO inhibitor tin protoporphyrin IX, supporting a role for HO-1 in the potentiation of the cytotoxic response. tin protoporphyrin IX 68-89 heme oxygenase 1 Homo sapiens 113-117 12433915-0 2003 Interaction of nitric oxide with human heme oxygenase-1. Nitric Oxide 15-27 heme oxygenase 1 Homo sapiens 39-55 12433915-2 2003 We have examined the binding of NO to human heme oxygenase-1 (hHO-1), an enzyme that oxidizes heme to biliverdin, CO, and free iron, to determine whether inhibition of hHO-1 by NO can contribute to the signaling interplay of NO and CO. An Fe(3+)-NO hHO-1-heme complex is formed with NO or the NO donors NOC9 or 2-(N,N-diethylamino)-diazenolate-2-oxide.sodium salt. Heme 44-48 heme oxygenase 1 Homo sapiens 62-67 12433915-2 2003 We have examined the binding of NO to human heme oxygenase-1 (hHO-1), an enzyme that oxidizes heme to biliverdin, CO, and free iron, to determine whether inhibition of hHO-1 by NO can contribute to the signaling interplay of NO and CO. An Fe(3+)-NO hHO-1-heme complex is formed with NO or the NO donors NOC9 or 2-(N,N-diethylamino)-diazenolate-2-oxide.sodium salt. Biliverdine 102-112 heme oxygenase 1 Homo sapiens 62-67 12433915-5 2003 The Fe(3+)-NO complex of hHO-1 is much more stable than that of met-Mb. fe(3+)-no 4-13 heme oxygenase 1 Homo sapiens 25-30 12433915-6 2003 Stopped-flow studies indicate that k(on) for formation of the hHO-1-heme Fe(3+)-NO complex is approximately 50-times faster, and k(off) 10 times slower, than for met-Mb, resulting in K(d) = 1.4 microm for NO. fe(3+)-no 73-82 heme oxygenase 1 Homo sapiens 62-67 14529548-3 2003 An inducible form of HO, HO-1, is induced by a variety of stresses such as oxidized lipoproteins, cytokines, hemodynamic changes, angiotensin II and nitric oxide (NO) in vascular wall. Nitric Oxide 149-161 heme oxygenase 1 Homo sapiens 25-29 14529548-5 2003 HO-1 induction in artery wall scavenges reactive oxygen species, which leads to the attenuation of monocyte adhesion and chemotaxis. Reactive Oxygen Species 40-63 heme oxygenase 1 Homo sapiens 0-4 14529549-1 2003 The heme oxygenase-1 (HO-1) enzyme catalyzes the rate-limiting reaction in the catabolism of heme yielding products with pleiotropic, but ultimately, cytoprotective activities. Heme 4-8 heme oxygenase 1 Homo sapiens 22-26 14529549-4 2003 Extensive analysis of the mouse gene, and to a lesser extent of the human gene, has identified a common mechanism the stress response element (StRE)/Nrf2 transcription factor pathway for gene regulation in response to a diverse array of HO-1 inducers including the substrate heme, various environmental and industrial toxins, and plant-derived polyphenolic compounds. Heme 275-279 heme oxygenase 1 Homo sapiens 237-241 14529549-4 2003 Extensive analysis of the mouse gene, and to a lesser extent of the human gene, has identified a common mechanism the stress response element (StRE)/Nrf2 transcription factor pathway for gene regulation in response to a diverse array of HO-1 inducers including the substrate heme, various environmental and industrial toxins, and plant-derived polyphenolic compounds. polyphenolic compounds 344-366 heme oxygenase 1 Homo sapiens 237-241 14529550-9 2003 Human HO-1 gene transfer into endothelial cells has been shown to attenuate Ang II- TNF- and heme-mediated DNA damage. Heme 93-97 heme oxygenase 1 Homo sapiens 6-10 14529552-3 2003 HO-1 is induced by nitric oxide (NO) in different biological systems and can control the increased production of this mediator observed in many inflammatory situations. Nitric Oxide 19-31 heme oxygenase 1 Homo sapiens 0-4 14529552-5 2003 Modulation of signal transduction pathways by HO-1 or products derived from its activity, such as carbon monoxide (CO), may mediate the anti-inflammatory effects of this protein. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 46-50 14529552-5 2003 Modulation of signal transduction pathways by HO-1 or products derived from its activity, such as carbon monoxide (CO), may mediate the anti-inflammatory effects of this protein. Carbon Monoxide 115-117 heme oxygenase 1 Homo sapiens 46-50 12498987-1 2003 In many models, a protective role for heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, has been demonstrated. Heme 38-42 heme oxygenase 1 Homo sapiens 56-60 12498987-5 2003 Disrupted HO-1 expression was associated with decreased lung reactive iron and iron-associated proteins, decreased NADPH cytochrome cp450 reductase activity, and decreased lung peroxidase activity compared to WT. Iron 70-74 heme oxygenase 1 Homo sapiens 10-14 12498987-5 2003 Disrupted HO-1 expression was associated with decreased lung reactive iron and iron-associated proteins, decreased NADPH cytochrome cp450 reductase activity, and decreased lung peroxidase activity compared to WT. Iron 79-83 heme oxygenase 1 Homo sapiens 10-14 12498987-7 2003 This suggests that disruption of HO-1 protects against hyperoxia by diminishing the generation of toxic reactive intermediates in the lung via iron and H(2)O(2). Iron 143-147 heme oxygenase 1 Homo sapiens 33-37 12498987-7 2003 This suggests that disruption of HO-1 protects against hyperoxia by diminishing the generation of toxic reactive intermediates in the lung via iron and H(2)O(2). Water 152-157 heme oxygenase 1 Homo sapiens 33-37 12469218-4 2003 Treatment of HLE/2E1 cells with succinylacetone (SA), a potent inhibitor of delta-aminolevulinate dehydratase and thereby heme synthesis, resulted in a further increase in ALAS-N mRNA but a decrease in HO-1 mRNA levels. succinylacetone 32-47 heme oxygenase 1 Homo sapiens 202-206 12469218-4 2003 Treatment of HLE/2E1 cells with succinylacetone (SA), a potent inhibitor of delta-aminolevulinate dehydratase and thereby heme synthesis, resulted in a further increase in ALAS-N mRNA but a decrease in HO-1 mRNA levels. succinylacetone 49-51 heme oxygenase 1 Homo sapiens 202-206 12469218-6 2003 These findings suggest that the overexpression of CYP2E1 results in the up-regulation of ALAS-N in order to meet with an increased demand for heme synthesis for CYP2E1 formation, while it also results in the up-regulation of HO-1 presumably by enzyme induction by free heme released from CYP2E1, which then results in the elimination of toxic excess free heme and ultimately restores the physiologic milieu. Heme 269-273 heme oxygenase 1 Homo sapiens 225-229 12469218-6 2003 These findings suggest that the overexpression of CYP2E1 results in the up-regulation of ALAS-N in order to meet with an increased demand for heme synthesis for CYP2E1 formation, while it also results in the up-regulation of HO-1 presumably by enzyme induction by free heme released from CYP2E1, which then results in the elimination of toxic excess free heme and ultimately restores the physiologic milieu. Heme 269-273 heme oxygenase 1 Homo sapiens 225-229 12598705-0 2003 The induction of heme oxygenase-1 by exogenous nitric oxide in ex vivo normal human skin. Nitric Oxide 47-59 heme oxygenase 1 Homo sapiens 17-33 12598705-3 2003 Many reports have revealed that exogenous nitric oxide (NO) can induce HO-1 in vitro, which is the induced isoform of HO. Nitric Oxide 42-54 heme oxygenase 1 Homo sapiens 71-75 12452690-0 2002 1H NMR detection of immobilized water molecules within a strong distal hydrogen-bonding network of substrate-bound human heme oxygenase-1. Hydrogen 0-2 heme oxygenase 1 Homo sapiens 121-137 12452690-0 2002 1H NMR detection of immobilized water molecules within a strong distal hydrogen-bonding network of substrate-bound human heme oxygenase-1. Water 32-37 heme oxygenase 1 Homo sapiens 121-137 12452690-0 2002 1H NMR detection of immobilized water molecules within a strong distal hydrogen-bonding network of substrate-bound human heme oxygenase-1. Hydrogen 71-79 heme oxygenase 1 Homo sapiens 121-137 12376363-5 2002 Actinomycin D and cycloheximide inhibited TGF-beta1-responsive HO-1 mRNA expression, indicating a requirement for transcription and de novo protein synthesis. Dactinomycin 0-13 heme oxygenase 1 Homo sapiens 63-67 12376366-8 2002 These results indicate that prolonged overexpression of HO-1 ultimately decreases sGC activity by limiting the availability of cellular heme. Heme 136-140 heme oxygenase 1 Homo sapiens 56-60 12376363-5 2002 Actinomycin D and cycloheximide inhibited TGF-beta1-responsive HO-1 mRNA expression, indicating a requirement for transcription and de novo protein synthesis. Cycloheximide 18-31 heme oxygenase 1 Homo sapiens 63-67 12376363-7 2002 A chemical inhibitor of p38 MAPK (SB-203580) abolished TGF-beta1-inducible HO-1 mRNA expression. SB 203580 34-43 heme oxygenase 1 Homo sapiens 75-79 12376363-8 2002 Both SB-203580 and expression of a dominant-negative mutant of p38 MAPK inhibited TGF-beta1-induced ho-1 gene activation, as assayed by luciferase activity of an ho-1 enhancer/luciferase fusion construct (pMHO1luc-33+SX2). SB 203580 5-14 heme oxygenase 1 Homo sapiens 100-104 12376366-0 2002 Modulation of cGMP by human HO-1 retrovirus gene transfer in pulmonary microvessel endothelial cells. Cyclic GMP 14-18 heme oxygenase 1 Homo sapiens 28-32 12376366-3 2002 Pulmonary cells that expressed hHO-1 exhibited a fourfold increase in HO activity associated with decreases in the steady-state levels of heme and cGMP without changes in soluble GC (sGC) and endothelial nitric oxide synthase (NOS) proteins or basal nitrite production. Heme 138-142 heme oxygenase 1 Homo sapiens 31-36 12376366-3 2002 Pulmonary cells that expressed hHO-1 exhibited a fourfold increase in HO activity associated with decreases in the steady-state levels of heme and cGMP without changes in soluble GC (sGC) and endothelial nitric oxide synthase (NOS) proteins or basal nitrite production. Cyclic GMP 147-151 heme oxygenase 1 Homo sapiens 31-36 12376366-3 2002 Pulmonary cells that expressed hHO-1 exhibited a fourfold increase in HO activity associated with decreases in the steady-state levels of heme and cGMP without changes in soluble GC (sGC) and endothelial nitric oxide synthase (NOS) proteins or basal nitrite production. Nitrites 250-257 heme oxygenase 1 Homo sapiens 31-36 12376366-4 2002 Heme elicited significant increases in CO production and intracellular cGMP levels in both pulmonary endothelial and pulmonary hHO-1-expressing cells. Heme 0-4 heme oxygenase 1 Homo sapiens 127-132 12376366-5 2002 N(omega)-nitro-L-arginine methyl ester (L-NAME), an inhibitor of NOS, significantly decreased cGMP levels in heme-treated pulmonary endothelial cells but not heme-treated hHO-1-expressing cells. Heme 109-113 heme oxygenase 1 Homo sapiens 171-176 12376366-6 2002 In the presence of exogenous heme, CO and cGMP levels in hHO-1-expressing cells exceeded the corresponding levels in pulmonary endothelial cells. Heme 29-33 heme oxygenase 1 Homo sapiens 57-62 12376366-6 2002 In the presence of exogenous heme, CO and cGMP levels in hHO-1-expressing cells exceeded the corresponding levels in pulmonary endothelial cells. Carbon Monoxide 35-37 heme oxygenase 1 Homo sapiens 57-62 12376366-6 2002 In the presence of exogenous heme, CO and cGMP levels in hHO-1-expressing cells exceeded the corresponding levels in pulmonary endothelial cells. Cyclic GMP 42-46 heme oxygenase 1 Homo sapiens 57-62 12396617-6 2002 However, inhibition of HO-1 after treatment with tin protoporphyrin reversed the immunomodulatory/antiapoptotic effects of Ad-IL-13 both in vivo (infectious transplantation tolerance), and in vitro (HUVECs). tin protoporphyrin IX 49-67 heme oxygenase 1 Homo sapiens 23-27 12394829-1 2002 The heme oxygenase-1 (HO-1) system, the rate-limiting step in the conversion of heme, is among the most critical of cytoprotective mechanisms activated during cellular stress. Heme 4-8 heme oxygenase 1 Homo sapiens 22-26 12394829-2 2002 The cytoprotection may result from the elimination of heme and the function of HO-1 downstream mediators, that is, biliverdin, carbon monoxide, and free iron. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 79-83 12394829-2 2002 The cytoprotection may result from the elimination of heme and the function of HO-1 downstream mediators, that is, biliverdin, carbon monoxide, and free iron. Carbon Monoxide 127-142 heme oxygenase 1 Homo sapiens 79-83 12394829-2 2002 The cytoprotection may result from the elimination of heme and the function of HO-1 downstream mediators, that is, biliverdin, carbon monoxide, and free iron. Iron 153-157 heme oxygenase 1 Homo sapiens 79-83 12352326-9 2002 Endothelial NOS mRNA was increased in cyclosporine-and tacrolimus-treated patients in comparison with controls (0.92 +/- 0.09, 0.96 +/- 0.04, and 0.37 +/- 0.05 respectively; p < 0.001), whereas no difference was found between patients and controls in HO-1 mRNA. Cyclosporine 38-50 heme oxygenase 1 Homo sapiens 254-258 12352326-9 2002 Endothelial NOS mRNA was increased in cyclosporine-and tacrolimus-treated patients in comparison with controls (0.92 +/- 0.09, 0.96 +/- 0.04, and 0.37 +/- 0.05 respectively; p < 0.001), whereas no difference was found between patients and controls in HO-1 mRNA. Tacrolimus 55-65 heme oxygenase 1 Homo sapiens 254-258 12352326-11 2002 Ramipril also reduced p22 (to 0.83 +/- 0.05 in cyclosporine, p < 0.03 and to 0.81 +/- 0.08 in tacrolimus; p < 0.01) and TGF-beta mRNA (to 0.72 +/- 01 in cyclosporine, p < 0.02, and to 0.73 +/- 0.05 in tacrolimus; p < 0.01) with no difference between groups, but it did not change HO-1 and ecNOS mRNA. Ramipril 0-8 heme oxygenase 1 Homo sapiens 292-296 12221210-5 2002 Histologic examination revealed that HO-1 was constitutively expressed in intestinal epithelial cells (IEC), and that glutamine increased the grade of HO-1 immunostaining (P </= 0.01). Glutamine 118-127 heme oxygenase 1 Homo sapiens 151-155 12382200-12 2002 Heme oxygenase 1 is essential for the catabolism of heme and in the recycling of hemoglobin iron in macrophages. Heme 52-56 heme oxygenase 1 Homo sapiens 0-16 12382200-12 2002 Heme oxygenase 1 is essential for the catabolism of heme and in the recycling of hemoglobin iron in macrophages. Iron 92-96 heme oxygenase 1 Homo sapiens 0-16 12352873-0 2002 Inhibition of ischemia/reperfusion injury and chronic graft deterioration by a single-donor treatment with cobalt-protoporphyrin for the induction of heme oxygenase-1. cobaltiprotoporphyrin 107-128 heme oxygenase 1 Homo sapiens 150-166 12352873-5 2002 Induction of HO-1 expression following cobalt-protoporphyrin (CoPP) treatment in organ donors prolonged graft survival and long-term function remarkably following extended periods of ischemia. cobaltiprotoporphyrin 39-60 heme oxygenase 1 Homo sapiens 13-17 12352873-5 2002 Induction of HO-1 expression following cobalt-protoporphyrin (CoPP) treatment in organ donors prolonged graft survival and long-term function remarkably following extended periods of ischemia. cobaltiprotoporphyrin 62-66 heme oxygenase 1 Homo sapiens 13-17 12207883-4 2002 The addition of SnMP (10 microM), an inhibitor of HO activity, to EC or SMC for 24h, resulted in significant abnormalities in DNA distribution and cell cycle progression compared to cells treated with the HO-1 inducers, heme (10 microM) or SnCl(2) (10 microM). tin mesoporphyrin 16-20 heme oxygenase 1 Homo sapiens 205-209 12221210-0 2002 Acute enteral glutamine infusion enhances heme oxygenase-1 expression in human duodenal mucosa. Glutamine 14-23 heme oxygenase 1 Homo sapiens 42-58 12221210-2 2002 Some experimental models suggest that induction of HO-1 by glutamine may contribute to the preservation of intestinal mucosa. Glutamine 59-68 heme oxygenase 1 Homo sapiens 51-55 12221210-3 2002 The effect of an enteral infusion of glutamine for 6 h on HO-1 expression in duodenal mucosa was studied in healthy men and women and compared with an isonitrogenous mixture of amino acids. Glutamine 37-46 heme oxygenase 1 Homo sapiens 58-62 12398878-3 2002 HO-1 expression was correlated with increased tissue iron and/or ferritin expression and increased inflammatory/oxidant load as measured by myeloperoxidase expression. Iron 53-57 heme oxygenase 1 Homo sapiens 0-4 12221210-6 2002 Glutamine also increased the percentage of HO-1 immunoreactive lamina propria cells (LPC, 10.5 vs. 7.5%, P </= 0.05). Glutamine 0-9 heme oxygenase 1 Homo sapiens 43-47 12221210-6 2002 Glutamine also increased the percentage of HO-1 immunoreactive lamina propria cells (LPC, 10.5 vs. 7.5%, P </= 0.05). lpc 85-88 heme oxygenase 1 Homo sapiens 43-47 12221210-7 2002 Glutamine significantly increased HO-1 mRNA expression compared with control amino acids: median (range) 156 (102-182) vs. 100 (68-179)%, P </= 0.05. Glutamine 0-9 heme oxygenase 1 Homo sapiens 34-38 12221210-9 2002 In conclusion, glutamine enhanced HO-1 mRNA and protein expression in human duodenal mucosa. Glutamine 15-24 heme oxygenase 1 Homo sapiens 34-38 12221210-10 2002 These data support further evaluation of the effects of glutamine on intestinal HO-1 during states of intestinal inflammation. Glutamine 56-65 heme oxygenase 1 Homo sapiens 80-84 12147246-6 2002 alpha-Naphthoflavone, which is known as an antagonist to 2,3,7,8-tetrachlorodibenzo-p-dioxin, inhibited the inductions of heme oxygenase 1, glutamate-cysteine ligase (modifier unit), and thioredoxin reductase by beta-naphthoflavone but not those of CYP1A1 and CYP1A2. alpha-naphthoflavone 0-20 heme oxygenase 1 Homo sapiens 122-138 12147246-6 2002 alpha-Naphthoflavone, which is known as an antagonist to 2,3,7,8-tetrachlorodibenzo-p-dioxin, inhibited the inductions of heme oxygenase 1, glutamate-cysteine ligase (modifier unit), and thioredoxin reductase by beta-naphthoflavone but not those of CYP1A1 and CYP1A2. Polychlorinated Dibenzodioxins 57-92 heme oxygenase 1 Homo sapiens 122-138 12147246-6 2002 alpha-Naphthoflavone, which is known as an antagonist to 2,3,7,8-tetrachlorodibenzo-p-dioxin, inhibited the inductions of heme oxygenase 1, glutamate-cysteine ligase (modifier unit), and thioredoxin reductase by beta-naphthoflavone but not those of CYP1A1 and CYP1A2. beta-Naphthoflavone 212-231 heme oxygenase 1 Homo sapiens 122-138 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Heme 61-65 heme oxygenase 1 Homo sapiens 18-22 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 0-16 12230868-3 2002 HO-1 is a cytoprotective enzyme that degrades heme, a potent oxidant, to generate carbon monoxide, biliverdin (subsequently reduced to bilirubin), and iron. Heme 46-50 heme oxygenase 1 Homo sapiens 0-4 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 18-22 12230868-3 2002 HO-1 is a cytoprotective enzyme that degrades heme, a potent oxidant, to generate carbon monoxide, biliverdin (subsequently reduced to bilirubin), and iron. Carbon Monoxide 82-97 heme oxygenase 1 Homo sapiens 0-4 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Iron 86-90 heme oxygenase 1 Homo sapiens 0-16 12230868-3 2002 HO-1 is a cytoprotective enzyme that degrades heme, a potent oxidant, to generate carbon monoxide, biliverdin (subsequently reduced to bilirubin), and iron. Biliverdine 99-109 heme oxygenase 1 Homo sapiens 0-4 12230868-3 2002 HO-1 is a cytoprotective enzyme that degrades heme, a potent oxidant, to generate carbon monoxide, biliverdin (subsequently reduced to bilirubin), and iron. Bilirubin 135-144 heme oxygenase 1 Homo sapiens 0-4 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Iron 86-90 heme oxygenase 1 Homo sapiens 18-22 12230868-3 2002 HO-1 is a cytoprotective enzyme that degrades heme, a potent oxidant, to generate carbon monoxide, biliverdin (subsequently reduced to bilirubin), and iron. Iron 151-155 heme oxygenase 1 Homo sapiens 0-4 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Biliverdine 101-111 heme oxygenase 1 Homo sapiens 0-16 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Biliverdine 101-111 heme oxygenase 1 Homo sapiens 18-22 12230869-0 2002 Effect of prostaglandin-J(2) on VEGF synthesis depends on the induction of heme oxygenase-1. prostaglandin-j 10-25 heme oxygenase 1 Homo sapiens 75-91 12230869-4 2002 Here we investigated the involvement of HO-1 in the 15d-PGJ(2)-mediated regulation of VEGF production by human microvascular endothelial cells (HMEC-1). 9-deoxy-delta-9-prostaglandin D2 56-62 heme oxygenase 1 Homo sapiens 40-44 12230869-1 2002 Heme oxygenase-1 (HO-1) is an inducible enzyme that degrades heme to carbon monoxide, iron ions, and biliverdin. Heme 61-65 heme oxygenase 1 Homo sapiens 0-16 12230869-8 2002 Activation of HO-1 with hemin or ectopic overexpression of HO-1 in HMEC-1 perfectly mimicked the effect of 15d-PGJ(2) and led to increased VEGF production. 15d-pgj 107-114 heme oxygenase 1 Homo sapiens 14-18 12230869-8 2002 Activation of HO-1 with hemin or ectopic overexpression of HO-1 in HMEC-1 perfectly mimicked the effect of 15d-PGJ(2) and led to increased VEGF production. 15d-pgj 107-114 heme oxygenase 1 Homo sapiens 59-63 12230869-9 2002 Importantly, the inhibition of the HO-1 pathway by tin protoporphyrin-IX significantly reduced the stimulatory effect of 15d-PGJ(2) on VEGF synthesis. tin protoporphyrin IX 51-72 heme oxygenase 1 Homo sapiens 35-39 12230869-9 2002 Importantly, the inhibition of the HO-1 pathway by tin protoporphyrin-IX significantly reduced the stimulatory effect of 15d-PGJ(2) on VEGF synthesis. 15d-pgj 121-128 heme oxygenase 1 Homo sapiens 35-39 12230871-3 2002 Here we present an overview of the heme degradation processes and relevant disorders by focusing on heme oxygenase-1 (HO-1), a key enzyme in heme catabolism. Heme 35-39 heme oxygenase 1 Homo sapiens 100-116 12230871-3 2002 Here we present an overview of the heme degradation processes and relevant disorders by focusing on heme oxygenase-1 (HO-1), a key enzyme in heme catabolism. Heme 35-39 heme oxygenase 1 Homo sapiens 118-122 12130498-2 2002 Deficiency of the heme-catabolizing enzyme, heme oxygenase-1 (HO-1), in both a human patient and transgenic knockout mice leads to an abundance of circulating heme and damage to vascular endothelium. Heme 18-22 heme oxygenase 1 Homo sapiens 44-60 12130498-2 2002 Deficiency of the heme-catabolizing enzyme, heme oxygenase-1 (HO-1), in both a human patient and transgenic knockout mice leads to an abundance of circulating heme and damage to vascular endothelium. Heme 18-22 heme oxygenase 1 Homo sapiens 62-66 12130498-2 2002 Deficiency of the heme-catabolizing enzyme, heme oxygenase-1 (HO-1), in both a human patient and transgenic knockout mice leads to an abundance of circulating heme and damage to vascular endothelium. Heme 44-48 heme oxygenase 1 Homo sapiens 62-66 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. Oxygen 74-80 heme oxygenase 1 Homo sapiens 0-4 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. tetrapyrrole biliverdin 101-124 heme oxygenase 1 Homo sapiens 0-4 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. Carbon Monoxide 126-141 heme oxygenase 1 Homo sapiens 0-4 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. Iron 155-159 heme oxygenase 1 Homo sapiens 0-4 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. Biliverdine 114-124 heme oxygenase 1 Homo sapiens 0-4 12151344-0 2002 Expression of interleukin-8, heme oxygenase-1 and vascular endothelial growth factor in DLD-1 colon carcinoma cells exposed to pyrrolidine dithiocarbamate. pyrrolidine dithiocarbamic acid 127-154 heme oxygenase 1 Homo sapiens 29-45 12230871-4 2002 HO-1 cleaves the porphyrin macrocycle of heme at the expense of molecular oxygen to release a linear tetrapyrrole biliverdin, carbon monoxide, and ferrous iron; biliverdin is rapidly reduced to bilirubin. Bilirubin 194-203 heme oxygenase 1 Homo sapiens 0-4 12230873-0 2002 Regulation of heme oxygenase-1 by redox signals involving nitric oxide. Nitric Oxide 58-70 heme oxygenase 1 Homo sapiens 14-30 12230873-2 2002 The importance of this protein in physiology and disease is underlined by the versatility of HO-1 inducers and the functional role attributed to HO-1 products (carbon monoxide and bilirubin) in conditions that are associated with moderate or severe cellular stress. Carbon Monoxide 160-175 heme oxygenase 1 Homo sapiens 145-149 12230873-2 2002 The importance of this protein in physiology and disease is underlined by the versatility of HO-1 inducers and the functional role attributed to HO-1 products (carbon monoxide and bilirubin) in conditions that are associated with moderate or severe cellular stress. Bilirubin 180-189 heme oxygenase 1 Homo sapiens 145-149 12230873-3 2002 An intriguing aspect is the recent evidence showing that nitric oxide, a ubiquitous signaling molecule, finely modulates the activation of HO-1 expression. Nitric Oxide 57-69 heme oxygenase 1 Homo sapiens 139-143 12230873-4 2002 As the effects of oxidative stress on the regulation of the HO-1 gene have been well established and characterized, this review will focus on the biological relevance of redox signals involving nitric oxide and reactive nitrogen species that lead to up-regulation of the HO-1 pathway, with particular emphasis on vascular tissues and the cardiovascular system. Nitric Oxide 194-206 heme oxygenase 1 Homo sapiens 60-64 12230873-4 2002 As the effects of oxidative stress on the regulation of the HO-1 gene have been well established and characterized, this review will focus on the biological relevance of redox signals involving nitric oxide and reactive nitrogen species that lead to up-regulation of the HO-1 pathway, with particular emphasis on vascular tissues and the cardiovascular system. Nitric Oxide 194-206 heme oxygenase 1 Homo sapiens 271-275 12230873-4 2002 As the effects of oxidative stress on the regulation of the HO-1 gene have been well established and characterized, this review will focus on the biological relevance of redox signals involving nitric oxide and reactive nitrogen species that lead to up-regulation of the HO-1 pathway, with particular emphasis on vascular tissues and the cardiovascular system. reactive nitrogen 211-228 heme oxygenase 1 Homo sapiens 60-64 12230873-4 2002 As the effects of oxidative stress on the regulation of the HO-1 gene have been well established and characterized, this review will focus on the biological relevance of redox signals involving nitric oxide and reactive nitrogen species that lead to up-regulation of the HO-1 pathway, with particular emphasis on vascular tissues and the cardiovascular system. reactive nitrogen 211-228 heme oxygenase 1 Homo sapiens 271-275 12222997-4 2002 An association of the relative increase (Delta, %) of these inflammatory markers with short (<25) (GT)(n) dinucleotide repeats in the HO-1 gene promoter was assessed. (n) dinucleotide 105-121 heme oxygenase 1 Homo sapiens 137-141 12227681-8 2002 Carvedilol increased plasma antioxidant power and HO-1 mRNA and reduced 3-nitrotyrosine and TGFbeta mRNA levels, while the same was not observed with nifedipine. Carvedilol 0-10 heme oxygenase 1 Homo sapiens 50-54 12176442-0 2002 Heme oxygenase-1 gene transfer prevents CD95/FasL-mediated apoptosis and improves liver allograft survival via carbon monoxide signaling pathway. Carbon Monoxide 111-126 heme oxygenase 1 Homo sapiens 0-16 12420741-0 2002 Management of oxidative stress by heme oxygenase-1 in cisplatin-induced toxicity in renal tubular cells. Cisplatin 54-63 heme oxygenase 1 Homo sapiens 34-50 12420741-1 2002 Induction of heme oxygenase-1 (HO-1) may serve as an immediate protective response during treatment with the cytostatic drug cisplatin (CDDP). Cisplatin 125-134 heme oxygenase 1 Homo sapiens 13-29 12420741-1 2002 Induction of heme oxygenase-1 (HO-1) may serve as an immediate protective response during treatment with the cytostatic drug cisplatin (CDDP). Cisplatin 136-140 heme oxygenase 1 Homo sapiens 13-29 12057767-10 2002 Indeed, beta-carotene or lycopene (0.5-1.0 microM) led to a further 1.5-fold rise in the UVA-induced HO-1 mRNA levels. beta Carotene 8-21 heme oxygenase 1 Homo sapiens 101-105 12082007-0 2002 Heme oxygenase-1-mediated protection: potential role of nonheme iron-nitric oxide complexes. Iron 64-68 heme oxygenase 1 Homo sapiens 0-16 12082007-0 2002 Heme oxygenase-1-mediated protection: potential role of nonheme iron-nitric oxide complexes. Nitric Oxide 69-81 heme oxygenase 1 Homo sapiens 0-16 12036874-0 2002 Heme oxygenase-1-derived carbon monoxide is an autocrine inhibitor of vascular smooth muscle cell growth. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 12036874-3 2002 The induction of HO-1 expression by serum was inhibited by actinomycin D or cycloheximide. Dactinomycin 59-72 heme oxygenase 1 Homo sapiens 17-21 12036874-3 2002 The induction of HO-1 expression by serum was inhibited by actinomycin D or cycloheximide. Cycloheximide 76-89 heme oxygenase 1 Homo sapiens 17-21 12057767-10 2002 Indeed, beta-carotene or lycopene (0.5-1.0 microM) led to a further 1.5-fold rise in the UVA-induced HO-1 mRNA levels. Lycopene 25-33 heme oxygenase 1 Homo sapiens 101-105 12118938-4 2002 However, the resistance of the HO-1 transfected VSMC against H2O2 was significantly raised. Hydrogen Peroxide 61-65 heme oxygenase 1 Homo sapiens 31-35 12118938-5 2002 This protective effect was dramatically diminished when the transfected VSMC were pretreated with ZnPP-IX, a specific inhibitor of HO, for 24 h. In addition, we found that the growth potential of the transfected cells was significantly inhibited directly by increased activity of HO-1, and this effect might be related to decreased phosphorylation of MAPK. zinc protoporphyrin 98-105 heme oxygenase 1 Homo sapiens 280-284 12182912-3 2002 A (GT)(n) dinucleotide repeat in the HO-1 gene promoter shows a length polymorphism that modulates the level of gene transcription. (n) dinucleotide 6-22 heme oxygenase 1 Homo sapiens 37-41 11880364-0 2002 Heme oxygenase-1-derived carbon monoxide requires the activation of transcription factor NF-kappa B to protect endothelial cells from tumor necrosis factor-alpha-mediated apoptosis. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 0-16 11880364-1 2002 We have shown that carbon monoxide (CO) generated by heme oxygenase-1 (HO-1) protects endothelial cells (EC) from tumor necrosis alpha (TNF-alpha)-mediated apoptosis. Carbon Monoxide 19-34 heme oxygenase 1 Homo sapiens 53-69 11880364-1 2002 We have shown that carbon monoxide (CO) generated by heme oxygenase-1 (HO-1) protects endothelial cells (EC) from tumor necrosis alpha (TNF-alpha)-mediated apoptosis. Carbon Monoxide 19-34 heme oxygenase 1 Homo sapiens 71-75 11880364-1 2002 We have shown that carbon monoxide (CO) generated by heme oxygenase-1 (HO-1) protects endothelial cells (EC) from tumor necrosis alpha (TNF-alpha)-mediated apoptosis. Carbon Monoxide 36-38 heme oxygenase 1 Homo sapiens 53-69 11880364-1 2002 We have shown that carbon monoxide (CO) generated by heme oxygenase-1 (HO-1) protects endothelial cells (EC) from tumor necrosis alpha (TNF-alpha)-mediated apoptosis. Carbon Monoxide 36-38 heme oxygenase 1 Homo sapiens 71-75 12166345-6 2002 These results suggest that HO-1 is up-regulated through heme-independent stimuli according to the development of portal hypertension, and that induced HO-1 plays a pathophysiological role in portal hypertension through carbon monoxide production. Carbon Monoxide 219-234 heme oxygenase 1 Homo sapiens 27-31 12166345-6 2002 These results suggest that HO-1 is up-regulated through heme-independent stimuli according to the development of portal hypertension, and that induced HO-1 plays a pathophysiological role in portal hypertension through carbon monoxide production. Carbon Monoxide 219-234 heme oxygenase 1 Homo sapiens 151-155 12028803-6 2002 Moreover, HO-1 expression was positively correlated to the higher organic carbon (OC) and polyaromatic hydrocarbons (PAHs) content of fine versus coarse PM, as well as the rise in PAH content that occurs in coarse PM during the winter months. Carbon 74-80 heme oxygenase 1 Homo sapiens 10-14 12028803-6 2002 Moreover, HO-1 expression was positively correlated to the higher organic carbon (OC) and polyaromatic hydrocarbons (PAHs) content of fine versus coarse PM, as well as the rise in PAH content that occurs in coarse PM during the winter months. polyaromatic hydrocarbons 90-115 heme oxygenase 1 Homo sapiens 10-14 12028803-6 2002 Moreover, HO-1 expression was positively correlated to the higher organic carbon (OC) and polyaromatic hydrocarbons (PAHs) content of fine versus coarse PM, as well as the rise in PAH content that occurs in coarse PM during the winter months. pahs 117-121 heme oxygenase 1 Homo sapiens 10-14 12028803-6 2002 Moreover, HO-1 expression was positively correlated to the higher organic carbon (OC) and polyaromatic hydrocarbons (PAHs) content of fine versus coarse PM, as well as the rise in PAH content that occurs in coarse PM during the winter months. p-Aminohippuric Acid 117-120 heme oxygenase 1 Homo sapiens 10-14 11773068-10 2002 The potential significance of the AP-1 binding is suggested by the finding that the response of HO-1, in COS cells stably transfected with antisense hBVR, with 66% reduced BVR activity, to superoxide anion (O(2)()) formed by menadione is attenuated, whereas induction by heme is not affected. carbonyl sulfide 105-108 heme oxygenase 1 Homo sapiens 96-100 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Carbon Monoxide 129-144 heme oxygenase 1 Homo sapiens 62-78 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Carbon Monoxide 129-144 heme oxygenase 1 Homo sapiens 80-84 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Carbon Monoxide 146-148 heme oxygenase 1 Homo sapiens 62-78 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Carbon Monoxide 146-148 heme oxygenase 1 Homo sapiens 80-84 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Bilirubin 154-163 heme oxygenase 1 Homo sapiens 62-78 12006180-4 2002 Hypoxia transiently increases the transcriptional rate of the heme oxygenase-1 (HO-1) gene, resulting in increased production of carbon monoxide (CO) and bilirubin. Bilirubin 154-163 heme oxygenase 1 Homo sapiens 80-84 12006183-0 2002 Modulation of endothelial cell apoptosis by heme oxygenase-1-derived carbon monoxide. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 44-60 12006183-3 2002 We have also shown that the antiapoptotic effect of HO-1 is mediated through heme catabolism into the gas carbon monoxide (CO). Heme 77-81 heme oxygenase 1 Homo sapiens 52-56 12006183-3 2002 We have also shown that the antiapoptotic effect of HO-1 is mediated through heme catabolism into the gas carbon monoxide (CO). Carbon Monoxide 106-121 heme oxygenase 1 Homo sapiens 52-56 12006183-3 2002 We have also shown that the antiapoptotic effect of HO-1 is mediated through heme catabolism into the gas carbon monoxide (CO). Carbon Monoxide 123-125 heme oxygenase 1 Homo sapiens 52-56 12042071-8 2002 Okadaic acid also induced heme oxygenase-1 mRNA in both cell lines, but the magnitude of induction was only twofold, and was rapid and transient. Okadaic Acid 0-12 heme oxygenase 1 Homo sapiens 26-42 12042071-9 2002 Okadaic acid and phorbol 12-myristate 13-acetate significantly decreased heme-mediated induction of heme oxygenase-1 mRNA in both Huh-7 and HepG2 cells. Okadaic Acid 0-12 heme oxygenase 1 Homo sapiens 100-116 12042071-9 2002 Okadaic acid and phorbol 12-myristate 13-acetate significantly decreased heme-mediated induction of heme oxygenase-1 mRNA in both Huh-7 and HepG2 cells. Tetradecanoylphorbol Acetate 17-48 heme oxygenase 1 Homo sapiens 100-116 12042071-9 2002 Okadaic acid and phorbol 12-myristate 13-acetate significantly decreased heme-mediated induction of heme oxygenase-1 mRNA in both Huh-7 and HepG2 cells. Heme 73-77 heme oxygenase 1 Homo sapiens 100-116 12042071-10 2002 Wortmannin diminished the heme-mediated induction of heme oxygenase-1 mRNA in HepG2 cells, but not Huh-7 cells. Wortmannin 0-10 heme oxygenase 1 Homo sapiens 53-69 12042071-10 2002 Wortmannin diminished the heme-mediated induction of heme oxygenase-1 mRNA in HepG2 cells, but not Huh-7 cells. Heme 26-30 heme oxygenase 1 Homo sapiens 53-69 12042074-1 2002 Heme-hemopexin coordinately regulates genes encoding protective proteins including metallothionein-I (MT-I) and heme oxygenase 1 (HO-1). Heme 0-4 heme oxygenase 1 Homo sapiens 112-128 12042074-1 2002 Heme-hemopexin coordinately regulates genes encoding protective proteins including metallothionein-I (MT-I) and heme oxygenase 1 (HO-1). Heme 0-4 heme oxygenase 1 Homo sapiens 130-134 12042074-2 2002 Hexamethylene-bisacetamide (HMBA), which induces differentiation and activates protein kinase C (PKC), synergistically augments the induction of both MT-I and MT-II mRNAs in response to heme-hemopexin, but attenuates the induction of HO-1. hexamethylene bisacetamide 0-26 heme oxygenase 1 Homo sapiens 234-238 12042074-2 2002 Hexamethylene-bisacetamide (HMBA), which induces differentiation and activates protein kinase C (PKC), synergistically augments the induction of both MT-I and MT-II mRNAs in response to heme-hemopexin, but attenuates the induction of HO-1. hexamethylene bisacetamide 28-32 heme oxygenase 1 Homo sapiens 234-238 12042074-2 2002 Hexamethylene-bisacetamide (HMBA), which induces differentiation and activates protein kinase C (PKC), synergistically augments the induction of both MT-I and MT-II mRNAs in response to heme-hemopexin, but attenuates the induction of HO-1. Heme 186-190 heme oxygenase 1 Homo sapiens 234-238 12042074-4 2002 Unlike the PKC-activating phorbol esters that induce MT-I and HO-1, HMBA has minimal effects on MT-I or HO-1. Phorbol Esters 26-40 heme oxygenase 1 Homo sapiens 62-66 12449018-6 2002 Finally, OxPAPC-induced expression of HO-1 was blocked by a platelet-activating factor (PAF) receptor antagonist. oxpapc 9-15 heme oxygenase 1 Homo sapiens 38-42 11773068-10 2002 The potential significance of the AP-1 binding is suggested by the finding that the response of HO-1, in COS cells stably transfected with antisense hBVR, with 66% reduced BVR activity, to superoxide anion (O(2)()) formed by menadione is attenuated, whereas induction by heme is not affected. Superoxides 189-205 heme oxygenase 1 Homo sapiens 96-100 11773068-10 2002 The potential significance of the AP-1 binding is suggested by the finding that the response of HO-1, in COS cells stably transfected with antisense hBVR, with 66% reduced BVR activity, to superoxide anion (O(2)()) formed by menadione is attenuated, whereas induction by heme is not affected. Superoxides 207-211 heme oxygenase 1 Homo sapiens 96-100 11773068-10 2002 The potential significance of the AP-1 binding is suggested by the finding that the response of HO-1, in COS cells stably transfected with antisense hBVR, with 66% reduced BVR activity, to superoxide anion (O(2)()) formed by menadione is attenuated, whereas induction by heme is not affected. Vitamin K 3 225-234 heme oxygenase 1 Homo sapiens 96-100 11773068-10 2002 The potential significance of the AP-1 binding is suggested by the finding that the response of HO-1, in COS cells stably transfected with antisense hBVR, with 66% reduced BVR activity, to superoxide anion (O(2)()) formed by menadione is attenuated, whereas induction by heme is not affected. Heme 271-275 heme oxygenase 1 Homo sapiens 96-100 11854143-7 2002 The levels of the CYP1A1 and CYP1B1 mRNAs induced by BAP were not significantly affected by coexposure to NaAsO(2); however, heme oxygenase 1 mRNA levels were markedly induced by coexposure to BAP and NaAsO(2). naaso 201-206 heme oxygenase 1 Homo sapiens 125-141 11854435-0 2002 Caffeic acid phenethyl ester and curcumin: a novel class of heme oxygenase-1 inducers. caffeic acid phenethyl ester 0-28 heme oxygenase 1 Homo sapiens 60-76 11854435-0 2002 Caffeic acid phenethyl ester and curcumin: a novel class of heme oxygenase-1 inducers. Curcumin 33-41 heme oxygenase 1 Homo sapiens 60-76 11854435-2 2002 Curcumin, a polyphenolic natural compound that possesses anti-tumor and anti-inflammatory properties, has been reported recently to induce potently HO-1 expression in vascular endothelial cells (Free Rad Biol Med 28:1303-1312, 2000). Curcumin 0-8 heme oxygenase 1 Homo sapiens 148-152 11854435-3 2002 Here, we extend our previous findings by showing that caffeic acid phenethyl ester (CAPE), another plant-derived phenolic agent, markedly increases heme oxygenase activity and HO-1 protein in astrocytes. caffeic acid phenethyl ester 54-82 heme oxygenase 1 Homo sapiens 176-180 11854435-3 2002 Here, we extend our previous findings by showing that caffeic acid phenethyl ester (CAPE), another plant-derived phenolic agent, markedly increases heme oxygenase activity and HO-1 protein in astrocytes. caffeic acid phenethyl ester 84-88 heme oxygenase 1 Homo sapiens 176-180 11854435-9 2002 These data suggest that regulation of HO-1 expression by polyphenolic compounds is evoked by a distinctive mechanism which is not necessarily linked to changes in glutathione but might depend on redox signals sustained by specific and targeted sulfydryl groups. Glutathione 163-174 heme oxygenase 1 Homo sapiens 38-42 11854435-9 2002 These data suggest that regulation of HO-1 expression by polyphenolic compounds is evoked by a distinctive mechanism which is not necessarily linked to changes in glutathione but might depend on redox signals sustained by specific and targeted sulfydryl groups. sulfydryl 244-253 heme oxygenase 1 Homo sapiens 38-42 11930234-1 2002 The heme oxygenase-1 (HO-1), a rate-limiting enzyme in heme metabolism, has been recently defined as a novel stress-stimulated protein, since the intracellular expression of HO-1 in response to various stimuli as oxidation, ischemia and endotoxin injury has been proved to be able to protect the cells from damage. Heme 4-8 heme oxygenase 1 Homo sapiens 174-178 11930234-5 2002 It was found that the HO-1 transfected-VSMCs presented dominant resistance to toxicity produced by exposure to H2O2, as a significant protective effect of HO-1 marked by cell survival and LDH leakage was observed when 200, 400 and 600 micromol/L of H2O2 were used. Hydrogen Peroxide 111-115 heme oxygenase 1 Homo sapiens 22-26 11930234-1 2002 The heme oxygenase-1 (HO-1), a rate-limiting enzyme in heme metabolism, has been recently defined as a novel stress-stimulated protein, since the intracellular expression of HO-1 in response to various stimuli as oxidation, ischemia and endotoxin injury has been proved to be able to protect the cells from damage. Heme 4-8 heme oxygenase 1 Homo sapiens 22-26 11930234-5 2002 It was found that the HO-1 transfected-VSMCs presented dominant resistance to toxicity produced by exposure to H2O2, as a significant protective effect of HO-1 marked by cell survival and LDH leakage was observed when 200, 400 and 600 micromol/L of H2O2 were used. Hydrogen Peroxide 111-115 heme oxygenase 1 Homo sapiens 155-159 11930234-5 2002 It was found that the HO-1 transfected-VSMCs presented dominant resistance to toxicity produced by exposure to H2O2, as a significant protective effect of HO-1 marked by cell survival and LDH leakage was observed when 200, 400 and 600 micromol/L of H2O2 were used. Hydrogen Peroxide 249-253 heme oxygenase 1 Homo sapiens 22-26 11930234-6 2002 The protection of HO-1 rapidly declined after the transfected-VSMCs were pretreated 24 h with an HO-1 specific inhibitor (ZnPP-IX). zinc protoporphyrin 122-129 heme oxygenase 1 Homo sapiens 18-22 11930234-6 2002 The protection of HO-1 rapidly declined after the transfected-VSMCs were pretreated 24 h with an HO-1 specific inhibitor (ZnPP-IX). zinc protoporphyrin 122-129 heme oxygenase 1 Homo sapiens 97-101 11930234-7 2002 The results of this investigation suggest that the functional expression of HO-1 gene within VSMCs raises an alternative ability to protect the vascular cells against active oxygen injury. Oxygen 174-180 heme oxygenase 1 Homo sapiens 76-80 11829463-11 2002 We conclude that an increase in endothelial cell HO-1 activity with subsequent generation of carbon monoxide, elicited by gene transfer, reversed the PDTC-mediated abnormalities in cell cycle progression and is thus a potential therapeutic means for attenuating the effects of oxidative stress-causing agents. Carbon Monoxide 93-108 heme oxygenase 1 Homo sapiens 49-53 11854317-2 2002 This study examined the induction of heme oxygenase-1 (HO-1), a carbon monoxide-generating inducible enzyme, in modulation of spermatogenesis. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 37-53 11820797-0 2002 Heme oxygenase-1 induction may explain the antioxidant profile of pentaerythrityl trinitrate. pentrinitrol 66-92 heme oxygenase 1 Homo sapiens 0-16 11820797-2 2002 In cultured endothelial cells derived from human umbilical vein, the active PETN metabolite PETriN (0.01-1 mM) increased heme oxygenase (HO)-1 mRNA and protein levels in a concentration-dependent fashion. Pentaerythritol Tetranitrate 76-80 heme oxygenase 1 Homo sapiens 121-142 11820797-2 2002 In cultured endothelial cells derived from human umbilical vein, the active PETN metabolite PETriN (0.01-1 mM) increased heme oxygenase (HO)-1 mRNA and protein levels in a concentration-dependent fashion. pentrinitrol 92-98 heme oxygenase 1 Homo sapiens 121-142 11854317-2 2002 This study examined the induction of heme oxygenase-1 (HO-1), a carbon monoxide-generating inducible enzyme, in modulation of spermatogenesis. Carbon Monoxide 64-79 heme oxygenase 1 Homo sapiens 55-59 11854317-4 2002 CdCl(2)-treated testes increased HO-1 activity and suppressed microsomal cytochromes P450, which are required for steroidogenesis. Cadmium Chloride 0-7 heme oxygenase 1 Homo sapiens 33-37 11854317-5 2002 CdCl(2)-elicited HO-1 occurred mostly in Leydig cells and coincided with CO generation, as judged by bilirubin-IXalpha immunoreactivity. Cadmium Chloride 0-7 heme oxygenase 1 Homo sapiens 17-21 11854317-5 2002 CdCl(2)-elicited HO-1 occurred mostly in Leydig cells and coincided with CO generation, as judged by bilirubin-IXalpha immunoreactivity. Bilirubin 101-110 heme oxygenase 1 Homo sapiens 17-21 11928711-3 2002 Exposure of mammalian cells to oxidative stimuli induces heme oxygenase-1 (HO-1), the rate-limiting enzyme in heme degradation, as well as the 32-kDa heat shock protein. Heme 57-61 heme oxygenase 1 Homo sapiens 75-79 12112002-2 2002 Here we show that NO donors including sodium nitroprusside (SNP) and spermine nonoate (SP-NO), and PGE(2) significantly stimulate HO-1 expression in RAW264.7 macrophages, associated with alternative induction on NO and PGE(2) in medium, respectively. Nitroprusside 38-58 heme oxygenase 1 Homo sapiens 130-134 11707454-2 2002 Depletion of nutrients results in cellular stress, which evokes adaptive and protective responses, one of which is the induction of heme oxygenase-1 (HO-1), a 32-kDa endoplasmic reticulum enzyme that catalyzes the rate-limiting step in heme degradation. Heme 132-136 heme oxygenase 1 Homo sapiens 150-154 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 44-51 heme oxygenase 1 Homo sapiens 89-93 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 44-51 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 187-194 heme oxygenase 1 Homo sapiens 89-93 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 187-194 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Fructose 307-315 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Galactose 317-326 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Mannose 328-335 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Deoxyglucose 341-355 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 187-194 heme oxygenase 1 Homo sapiens 89-93 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. Glucose 187-194 heme oxygenase 1 Homo sapiens 218-222 11707454-3 2002 Incubation of HepG2 human hepatoma cells in glucose-free medium resulted in an increased HO-1 mRNA content, reaching a maximum of approximately 25-fold over control cells after 12 h. The glucose-dependent induction of HO-1 mRNA was concentration-dependent (k(12) approximately 0.5 mm) and was attenuated by fructose, galactose, mannose, and 2-deoxyglucose, but not by the non-metabolizable glucose analog, 3-O-methylglucose. 3-O-Methylglucose 406-423 heme oxygenase 1 Homo sapiens 218-222 11707454-5 2002 These results demonstrate that glucose availability regulates transcription of the HO-1 gene via a pathway that is different from the unfolded protein response. Glucose 31-38 heme oxygenase 1 Homo sapiens 83-87 12489116-3 2002 Using treatment of cell cultures with hyperbaric oxygen (HBO) as a model for oxidative stress, we have shown an induction of HO-1 in isolated human lymphocytes after a single HBO exposure and protection of these cells against DNA damage by subsequent oxidative stress. Oxygen 49-55 heme oxygenase 1 Homo sapiens 125-129 11948696-4 2002 In cells transduced by HOP-driven HO-1 gene, there was a decrease in basal cyclooxygenase (COX) activity as measured by PGE(2). Prostaglandins E 120-123 heme oxygenase 1 Homo sapiens 34-38 11948696-5 2002 The degree of HO-1 expression and, consequently, the levels of cellular heme were directly related to COX activity. Heme 72-76 heme oxygenase 1 Homo sapiens 14-18 12112002-6 2002 And, NSAIDs aspirin and diclofenase dose dependently inhibited LPS/IFN-gamma-induced HO-1 protein accompanied by suppression of PGE(2) (not NO) production. diclofenase 24-35 heme oxygenase 1 Homo sapiens 85-89 12112002-7 2002 PD98059 (a specific inhibitor of MEKK), but not SB203580 (a specific inhibitor of p38 kinase), attenuated PGE(2) (not SP-NO) induced HO-1 protein. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 0-7 heme oxygenase 1 Homo sapiens 133-137 12112002-7 2002 PD98059 (a specific inhibitor of MEKK), but not SB203580 (a specific inhibitor of p38 kinase), attenuated PGE(2) (not SP-NO) induced HO-1 protein. Prostaglandins E 106-109 heme oxygenase 1 Homo sapiens 133-137 12112002-8 2002 Under UVC (100 J/m(2)) and UVB (50 J/m(2)) irradiation, PGE(2) or SP-NO treatment prevents cells from UVC or UVB-induced cell death, and HO-1 inhibitor tin protoporphyrin (SnPP) reverses the preventive effects of PGE(2) and SP-NO. Prostaglandins E 56-59 heme oxygenase 1 Homo sapiens 137-141 12112002-8 2002 Under UVC (100 J/m(2)) and UVB (50 J/m(2)) irradiation, PGE(2) or SP-NO treatment prevents cells from UVC or UVB-induced cell death, and HO-1 inhibitor tin protoporphyrin (SnPP) reverses the preventive effects of PGE(2) and SP-NO. sp-no 66-71 heme oxygenase 1 Homo sapiens 137-141 12112002-8 2002 Under UVC (100 J/m(2)) and UVB (50 J/m(2)) irradiation, PGE(2) or SP-NO treatment prevents cells from UVC or UVB-induced cell death, and HO-1 inhibitor tin protoporphyrin (SnPP) reverses the preventive effects of PGE(2) and SP-NO. tin protoporphyrin IX 152-170 heme oxygenase 1 Homo sapiens 137-141 12112002-10 2002 These results demonstrated that inflammatory molecules NO and PGE(2) were potent inducers of HO-1 gene, and protected cells from UV-irradiation-induced cell death through HO-1 induction. Prostaglandins E 62-65 heme oxygenase 1 Homo sapiens 93-97 12112002-10 2002 These results demonstrated that inflammatory molecules NO and PGE(2) were potent inducers of HO-1 gene, and protected cells from UV-irradiation-induced cell death through HO-1 induction. Prostaglandins E 62-65 heme oxygenase 1 Homo sapiens 171-175 12112002-2 2002 Here we show that NO donors including sodium nitroprusside (SNP) and spermine nonoate (SP-NO), and PGE(2) significantly stimulate HO-1 expression in RAW264.7 macrophages, associated with alternative induction on NO and PGE(2) in medium, respectively. spermine nitric oxide complex 69-85 heme oxygenase 1 Homo sapiens 130-134 12112002-2 2002 Here we show that NO donors including sodium nitroprusside (SNP) and spermine nonoate (SP-NO), and PGE(2) significantly stimulate HO-1 expression in RAW264.7 macrophages, associated with alternative induction on NO and PGE(2) in medium, respectively. sp-no 87-92 heme oxygenase 1 Homo sapiens 130-134 12112002-2 2002 Here we show that NO donors including sodium nitroprusside (SNP) and spermine nonoate (SP-NO), and PGE(2) significantly stimulate HO-1 expression in RAW264.7 macrophages, associated with alternative induction on NO and PGE(2) in medium, respectively. Prostaglandins E 99-102 heme oxygenase 1 Homo sapiens 130-134 12112002-4 2002 In the presence of lipopolysaccharide and interferon-gamma (LPS/IFN-gamma), HO-1 protein was induced slightly but significantly, and SNP, SP-NO, and PGE(2) enhanced HO-1 protein induced by LPS/IFN-gamma. sp-no 138-143 heme oxygenase 1 Homo sapiens 165-169 12112002-4 2002 In the presence of lipopolysaccharide and interferon-gamma (LPS/IFN-gamma), HO-1 protein was induced slightly but significantly, and SNP, SP-NO, and PGE(2) enhanced HO-1 protein induced by LPS/IFN-gamma. Prostaglandins E 149-152 heme oxygenase 1 Homo sapiens 165-169 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. Arginine 0-10 heme oxygenase 1 Homo sapiens 109-113 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. n-nitro-l-arginine methyl ester 19-50 heme oxygenase 1 Homo sapiens 109-113 12397597-5 2002 Endothelial cells stably transfected with the retrovirus containing the human HO-1 gene exhibited a several-fold increase in HO-1 protein levels, which was accompanied by an increase in HO activity and a marked decrease in PGE(2) and 6-keto PGF(1alpha) levels. Prostaglandins E 223-226 heme oxygenase 1 Homo sapiens 78-82 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. NG-Nitroarginine Methyl Ester 52-58 heme oxygenase 1 Homo sapiens 109-113 12397597-5 2002 Endothelial cells stably transfected with the retrovirus containing the human HO-1 gene exhibited a several-fold increase in HO-1 protein levels, which was accompanied by an increase in HO activity and a marked decrease in PGE(2) and 6-keto PGF(1alpha) levels. 6-keto pgf 234-244 heme oxygenase 1 Homo sapiens 78-82 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. Nitroarginine 19-37 heme oxygenase 1 Homo sapiens 109-113 12397597-5 2002 Endothelial cells stably transfected with the retrovirus containing the human HO-1 gene exhibited a several-fold increase in HO-1 protein levels, which was accompanied by an increase in HO activity and a marked decrease in PGE(2) and 6-keto PGF(1alpha) levels. 6-keto pgf 234-244 heme oxygenase 1 Homo sapiens 125-129 12397597-10 2002 The levels of PGI(2) decreased in cells transduced with HO-1 sense and increased in cells transduced with HO-1 in antisense orientation. Epoprostenol 14-20 heme oxygenase 1 Homo sapiens 56-60 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. Nitroarginine 84-87 heme oxygenase 1 Homo sapiens 109-113 12397597-10 2002 The levels of PGI(2) decreased in cells transduced with HO-1 sense and increased in cells transduced with HO-1 in antisense orientation. Epoprostenol 14-20 heme oxygenase 1 Homo sapiens 106-110 12112002-5 2002 L-Arginine analogs N-nitro-L-arginine methyl ester (L-NAME) and N-nitro-L-arginine (NLA) significantly block HO-1 protein induced by LPS/IFN-gamma associated with a decrease in NO (not PGE(2)) production. Prostaglandins E 185-188 heme oxygenase 1 Homo sapiens 109-113 12397597-13 2002 Tin mesoporphyrin (SnMP) reversed the protective effect of HO-1. tin mesoporphyrin 0-17 heme oxygenase 1 Homo sapiens 59-63 12397597-13 2002 Tin mesoporphyrin (SnMP) reversed the protective effect of HO-1. tin mesoporphyrin 19-23 heme oxygenase 1 Homo sapiens 59-63 12112002-6 2002 And, NSAIDs aspirin and diclofenase dose dependently inhibited LPS/IFN-gamma-induced HO-1 protein accompanied by suppression of PGE(2) (not NO) production. Aspirin 12-19 heme oxygenase 1 Homo sapiens 85-89 11925914-8 2002 Third, we demonstrated that exhaled carbon monoxide concentrations caused by heme oxygenase-1 upregulation, may be a useful noninvasive means of monitoring airway inflammation and of controlling elderly patients with bronchial asthma. Carbon Monoxide 36-51 heme oxygenase 1 Homo sapiens 77-93 11752115-5 2002 Endothelial cells stably transfected with the human HO-1 gene exhibited a severalfold increase in human HO-1 mRNA levels, which was accompanied by an increase in HO activity and a marked decrease in prostaglandin (PG) E(2) and 6-keto-PGF(1alpha) levels. Prostaglandins E 199-219 heme oxygenase 1 Homo sapiens 52-56 11752115-5 2002 Endothelial cells stably transfected with the human HO-1 gene exhibited a severalfold increase in human HO-1 mRNA levels, which was accompanied by an increase in HO activity and a marked decrease in prostaglandin (PG) E(2) and 6-keto-PGF(1alpha) levels. Prostaglandins E 199-219 heme oxygenase 1 Homo sapiens 104-108 11752115-5 2002 Endothelial cells stably transfected with the human HO-1 gene exhibited a severalfold increase in human HO-1 mRNA levels, which was accompanied by an increase in HO activity and a marked decrease in prostaglandin (PG) E(2) and 6-keto-PGF(1alpha) levels. 6-keto-pgf 227-237 heme oxygenase 1 Homo sapiens 52-56 11752115-5 2002 Endothelial cells stably transfected with the human HO-1 gene exhibited a severalfold increase in human HO-1 mRNA levels, which was accompanied by an increase in HO activity and a marked decrease in prostaglandin (PG) E(2) and 6-keto-PGF(1alpha) levels. 6-keto-pgf 227-237 heme oxygenase 1 Homo sapiens 104-108 11752115-6 2002 Exposure of cells to CoCl(2), an inducer of HO-1 gene expression, resulted in increases in HO-1 protein levels and HO activity. cobaltous chloride 21-28 heme oxygenase 1 Homo sapiens 44-48 11752115-6 2002 Exposure of cells to CoCl(2), an inducer of HO-1 gene expression, resulted in increases in HO-1 protein levels and HO activity. cobaltous chloride 21-28 heme oxygenase 1 Homo sapiens 91-95 11752115-9 2002 The degree of HO-1 expression and, consequently, the level of cellular heme, were directly related to COX activity. Heme 71-75 heme oxygenase 1 Homo sapiens 14-18 15499991-3 2002 Three isoforms of heme oxygenase (HO) have been described: two constitutively expressed isoforms, HO-2 and HO-3, and an inducible isoform, HO-1 that is increased as an adaptive response to several injurious stimuli including heme, hyperoxia, hypoxia, endotoxin and heavy metals. Heme 18-22 heme oxygenase 1 Homo sapiens 139-143 11592943-0 2001 Mechanism of heme oxygenase-1 gene induction by curcumin in human renal proximal tubule cells. Curcumin 48-56 heme oxygenase 1 Homo sapiens 13-29 11741586-0 2001 The effect of beta-carotene on the expression of interleukin-6 and heme oxygenase-1 in UV-irradiated human skin fibroblasts in vitro. beta Carotene 14-27 heme oxygenase 1 Homo sapiens 67-83 11741586-2 2001 However, we recently demonstrated that beta-carotene has a pro-oxidant potential in cultured human skin fibroblasts because it enhances the UVA induction of heme oxygenase-1 (HO-1). beta Carotene 39-52 heme oxygenase 1 Homo sapiens 157-173 11741586-2 2001 However, we recently demonstrated that beta-carotene has a pro-oxidant potential in cultured human skin fibroblasts because it enhances the UVA induction of heme oxygenase-1 (HO-1). beta Carotene 39-52 heme oxygenase 1 Homo sapiens 175-179 11741586-4 2001 Singlet oxygen quencher sodium azide abrogated up-regulation of IL-6, and likewise also of HO-1. Sodium Azide 24-36 heme oxygenase 1 Homo sapiens 91-95 11592943-5 2001 Surprisingly, curcumin by itself was a very potent inducer of HO-1. Curcumin 14-22 heme oxygenase 1 Homo sapiens 62-66 11592943-7 2001 To evaluate the mechanism of curcumin-mediated induction of HO-1, confluent human renal proximal tubule cells were exposed to curcumin (1-8 microM). Curcumin 29-37 heme oxygenase 1 Homo sapiens 60-64 11751428-0 2001 Involvement of heme oxygenase-1 (HO-1) in the adaptive protection of human lymphocytes after hyperbaric oxygen (HBO) treatment. Oxygen 20-26 heme oxygenase 1 Homo sapiens 33-37 11751428-2 2001 Our previous studies on hyperbaric oxygen (HBO; i.e. exposure to pure oxygen under high ambient pressure) indicated clearly increased levels of HO-1 in lymphocytes of volunteers 24 h after HBO treatment (1 h at 1.5 bar). Oxygen 35-41 heme oxygenase 1 Homo sapiens 144-148 11751428-2 2001 Our previous studies on hyperbaric oxygen (HBO; i.e. exposure to pure oxygen under high ambient pressure) indicated clearly increased levels of HO-1 in lymphocytes of volunteers 24 h after HBO treatment (1 h at 1.5 bar). Oxygen 70-76 heme oxygenase 1 Homo sapiens 144-148 11751428-8 2001 Treatment with the specific HO-1 inhibitor tin-mesoporphyrin IX (SnMP) led to a complete abrogation of HBO-induced adaptive protection in human lymphocytes. tin-mesoporphyrin ix 43-63 heme oxygenase 1 Homo sapiens 28-32 11751428-8 2001 Treatment with the specific HO-1 inhibitor tin-mesoporphyrin IX (SnMP) led to a complete abrogation of HBO-induced adaptive protection in human lymphocytes. tin mesoporphyrin 65-69 heme oxygenase 1 Homo sapiens 28-32 11728461-0 2001 Homocysteine attenuates endothelial haem oxygenase-1 induction by nitric oxide (NO) and hypoxia. Homocysteine 0-12 heme oxygenase 1 Homo sapiens 36-52 11728461-0 2001 Homocysteine attenuates endothelial haem oxygenase-1 induction by nitric oxide (NO) and hypoxia. Nitric Oxide 66-78 heme oxygenase 1 Homo sapiens 36-52 11728461-2 2001 Here, we examined how Hcy modulates the induction of the stress protein haem oxygenase-1 (HO-1) evoked by NO releasing agents and hypoxia in vascular endothelial cells. Homocysteine 22-25 heme oxygenase 1 Homo sapiens 72-88 11728461-2 2001 Here, we examined how Hcy modulates the induction of the stress protein haem oxygenase-1 (HO-1) evoked by NO releasing agents and hypoxia in vascular endothelial cells. Homocysteine 22-25 heme oxygenase 1 Homo sapiens 90-94 11728461-3 2001 We found that Hcy (0.5 mM) markedly reduced the increase in haem oxygenase activity and HO-1 protein expression induced by sodium nitroprusside (SNP, 0.5 mM) but did not affect HO-1 activation mediated by S-nitroso-N-acetyl-penicillamine. Homocysteine 14-17 heme oxygenase 1 Homo sapiens 88-92 11728461-3 2001 We found that Hcy (0.5 mM) markedly reduced the increase in haem oxygenase activity and HO-1 protein expression induced by sodium nitroprusside (SNP, 0.5 mM) but did not affect HO-1 activation mediated by S-nitroso-N-acetyl-penicillamine. Nitroprusside 123-143 heme oxygenase 1 Homo sapiens 88-92 11728461-5 2001 Interestingly, high levels of Hcy were also able to abolish hypoxia-mediated HO-1 expression in a concentration-dependent manner. Homocysteine 30-33 heme oxygenase 1 Homo sapiens 77-81 11707736-5 2001 Increased HO-1 protein was associated with a brisk and early increase in catalytically active iron and a robust increase in cellular ferritin. Iron 94-98 heme oxygenase 1 Homo sapiens 10-14 11592943-9 2001 Coincubation of curcumin with actinomycin D completely blocked the upregulation of HO-1 mRNA. Curcumin 16-24 heme oxygenase 1 Homo sapiens 83-87 11592943-9 2001 Coincubation of curcumin with actinomycin D completely blocked the upregulation of HO-1 mRNA. Dactinomycin 30-43 heme oxygenase 1 Homo sapiens 83-87 11592943-10 2001 Blockade of nuclear factor-kappaB (NF-kappaB) with an IkappaBalpha phosphorylation inhibitor attenuated curcumin-mediated induction of HO-1 mRNA and protein. Curcumin 104-112 heme oxygenase 1 Homo sapiens 135-139 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Heme 60-64 heme oxygenase 1 Homo sapiens 0-16 11592943-11 2001 These data demonstrate that curcumin induces HO-1 mRNA and protein in renal proximal tubule cells. Curcumin 28-36 heme oxygenase 1 Homo sapiens 45-49 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Heme 60-64 heme oxygenase 1 Homo sapiens 18-22 11592943-12 2001 HO-1 induction by curcumin is mediated, at least in part, via transcriptional mechanisms and involves the NF-kappaB pathway. Curcumin 18-26 heme oxygenase 1 Homo sapiens 0-4 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Iron 88-92 heme oxygenase 1 Homo sapiens 0-16 11592948-2 2001 By Northern blot analysis, HO-1 mRNA expression was found to significantly increase in response to reduction of PO(2) in culture medium. PO-2 112-117 heme oxygenase 1 Homo sapiens 27-31 11592948-4 2001 This hypoxia-induced upregulation of HO-1 mRNA was significantly blocked by HIF-1alpha inhibition with ferrous ammonium sulfate. ammonium ferrous sulfate 103-127 heme oxygenase 1 Homo sapiens 37-41 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Heme 63-67 heme oxygenase 1 Homo sapiens 0-16 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Iron 88-92 heme oxygenase 1 Homo sapiens 18-22 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Heme 63-67 heme oxygenase 1 Homo sapiens 18-22 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Carbon Monoxide 71-86 heme oxygenase 1 Homo sapiens 0-16 11592948-6 2001 Both desferrioxamine and CoCl(2) markedly increased HIF-1alpha mRNA and protein levels and resulted in the upregulation of HO-1 mRNA but not HO-2. Deferoxamine 5-20 heme oxygenase 1 Homo sapiens 123-127 11592948-6 2001 Both desferrioxamine and CoCl(2) markedly increased HIF-1alpha mRNA and protein levels and resulted in the upregulation of HO-1 mRNA but not HO-2. cobaltous chloride 25-32 heme oxygenase 1 Homo sapiens 123-127 11592948-7 2001 Furthermore, inhibition of HIF-1alpha degradation by CBZ-LLL, an inhibitor of ubiquitin-proteasome, significantly increased HIF-1alpha protein and HO-1 mRNA but not HO-2 in these cells. carbobenzoxy-leucyl-leucyl-leucine 53-60 heme oxygenase 1 Homo sapiens 147-151 11592948-8 2001 Using cis-element oligodeoxynucleotide transfection to specifically decoy HIF-1alpha and block HIF-1alpha binding, increased mRNA expression of HO-1 in response to hypoxia and CoCl(2) was attenuated. cobaltous chloride 176-183 heme oxygenase 1 Homo sapiens 144-148 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Carbon Monoxide 71-86 heme oxygenase 1 Homo sapiens 18-22 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Bilirubin 88-97 heme oxygenase 1 Homo sapiens 0-16 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Bilirubin 88-97 heme oxygenase 1 Homo sapiens 18-22 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Iron 103-107 heme oxygenase 1 Homo sapiens 0-16 11668058-1 2001 Heme oxygenase-1 (HO-1) catalyzes the enzymatic degradation of heme to carbon monoxide, bilirubin, and iron. Iron 103-107 heme oxygenase 1 Homo sapiens 18-22 11668058-3 2001 Here, we investigated the effect of severe hypoxia and reoxygenation on HO-1 expression in cardiomyocytes and determined whether HO-1 and its product, bilirubin, have a protective role against reoxygenation damage. Bilirubin 151-160 heme oxygenase 1 Homo sapiens 129-133 11668058-9 2001 These results indicate that the HO-1-bilirubin pathway can effectively defend hypoxic cardiomyocytes against reoxygenation injury and highlight the issue of heme availability in the cytoprotective action afforded by HO-1. Bilirubin 37-46 heme oxygenase 1 Homo sapiens 32-36 11668058-9 2001 These results indicate that the HO-1-bilirubin pathway can effectively defend hypoxic cardiomyocytes against reoxygenation injury and highlight the issue of heme availability in the cytoprotective action afforded by HO-1. Bilirubin 37-46 heme oxygenase 1 Homo sapiens 216-220 11668058-9 2001 These results indicate that the HO-1-bilirubin pathway can effectively defend hypoxic cardiomyocytes against reoxygenation injury and highlight the issue of heme availability in the cytoprotective action afforded by HO-1. Heme 157-161 heme oxygenase 1 Homo sapiens 216-220 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 0-16 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Carbon Monoxide 94-109 heme oxygenase 1 Homo sapiens 18-22 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 0-16 11592943-1 2001 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme degradation, releasing iron, carbon monoxide, and biliverdin. Biliverdine 115-125 heme oxygenase 1 Homo sapiens 18-22 11593038-5 2001 Overexpression of HO-1 AS was associated with a long-term decrease (45%) of endogenous HO-1 protein and an increase (167%) in unmetabolized exogenous heme in HMEC-1 cells. Heme 150-154 heme oxygenase 1 Homo sapiens 18-22 11593038-6 2001 Carbon monoxide (CO) production in HO-1 S- or AS-transduced HMEC-1 cells after heme treatment was increased (159%) or decreased (50%), respectively, compared with nontransduced cells. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 35-39 11593038-6 2001 Carbon monoxide (CO) production in HO-1 S- or AS-transduced HMEC-1 cells after heme treatment was increased (159%) or decreased (50%), respectively, compared with nontransduced cells. Carbon Monoxide 17-19 heme oxygenase 1 Homo sapiens 35-39 11593038-6 2001 Carbon monoxide (CO) production in HO-1 S- or AS-transduced HMEC-1 cells after heme treatment was increased (159%) or decreased (50%), respectively, compared with nontransduced cells. Heme 79-83 heme oxygenase 1 Homo sapiens 35-39 11600581-6 2001 The detection of mRNA and protein for HO-1 and HO-2 in normal human endometrium suggests that the carbon monoxide/HO system may play a role in the local control of endometrial function. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 38-51 11581219-6 2001 Differentiated ARPE-19 cells treated with tBH or H2O2 resulted in upregulation of the HO-1 and FGFR1 transcripts. tert-Butylhydroperoxide 42-45 heme oxygenase 1 Homo sapiens 86-90 11581219-6 2001 Differentiated ARPE-19 cells treated with tBH or H2O2 resulted in upregulation of the HO-1 and FGFR1 transcripts. Hydrogen Peroxide 49-53 heme oxygenase 1 Homo sapiens 86-90 11572959-7 2001 Macrophage-inducible NOS generates NO to kill other cells, whereas HO1 generates bilirubin to exert antioxidant cytoprotective effects and also provides cytoprotection by facilitating iron extrusion from cells. Bilirubin 81-90 heme oxygenase 1 Homo sapiens 67-70 11571246-3 2001 METHODS AND RESULTS: Infection of rat aortic smooth muscle cells with adenovirus carrying the human HO-1 gene (Adv-HO-1) resulted in a high-level expression of HO-1 protein, which effectively reduced the hemin-induced iron overload in these cells. Hemin 204-209 heme oxygenase 1 Homo sapiens 100-104 11718398-1 2001 PURPOSE: To determine if an association exists between postdilation restenosis and heme oxygenase-1 (HO-1), which is induced by balloon injury and inhibits neointimal formation through the action of endogenous carbon monoxide. Carbon Monoxide 210-225 heme oxygenase 1 Homo sapiens 83-99 11718398-1 2001 PURPOSE: To determine if an association exists between postdilation restenosis and heme oxygenase-1 (HO-1), which is induced by balloon injury and inhibits neointimal formation through the action of endogenous carbon monoxide. Carbon Monoxide 210-225 heme oxygenase 1 Homo sapiens 101-105 11718398-2 2001 A dinucleotide repeat in the promoter region of the HO-1 gene shows a length polymorphism that modulates the level of gene transcription. Dinucleoside Phosphates 2-14 heme oxygenase 1 Homo sapiens 52-56 11718398-7 2001 Patients with short (<25 GT) dinucleotide repeats in the HO-1 gene promoter on either allele had restenosis significantly less often than patients with longer (> or = 25 GT) dinucleotide repeats (p = 0.01). Dinucleoside Phosphates 32-44 heme oxygenase 1 Homo sapiens 60-64 11768235-0 2001 Induction of heme oxygenase-1 by phenylarsine oxide. oxophenylarsine 33-51 heme oxygenase 1 Homo sapiens 13-29 11768235-2 2001 Heme oxygenase-1, the major inducible isoform of heme oxygenase (HO), can be induced by heme and numerous other physical and chemical factors, many of which cause cellular "stress". Heme 49-53 heme oxygenase 1 Homo sapiens 0-16 11768235-5 2001 We hypothesized that phenylarsine oxide (PAO), an inhibitor of protein tyrosine phosphatases (PTPs), might up-regulate the HO-1 gene. oxophenylarsine 21-39 heme oxygenase 1 Homo sapiens 123-127 11768235-5 2001 We hypothesized that phenylarsine oxide (PAO), an inhibitor of protein tyrosine phosphatases (PTPs), might up-regulate the HO-1 gene. oxophenylarsine 41-44 heme oxygenase 1 Homo sapiens 123-127 11768235-6 2001 Here, we show that a remarkably brief (1-15 min) exposure of normal hepatocytes to low concentrations (0.5-3 microM) of PAO produces a marked increase in mRNA and protein of HO-1. oxophenylarsine 120-123 heme oxygenase 1 Homo sapiens 174-178 11768235-9 2001 Addition of thiol donors abrogated the PAO-mediated induction of HO-1 in a dose dependent fashion. Sulfhydryl Compounds 12-17 heme oxygenase 1 Homo sapiens 65-69 11768235-9 2001 Addition of thiol donors abrogated the PAO-mediated induction of HO-1 in a dose dependent fashion. oxophenylarsine 39-42 heme oxygenase 1 Homo sapiens 65-69 11768235-12 2001 Our results are consistent with the proposition that induction of HO-1 by PAO involves inhibition of specific PTP(s), and that the mechanisms of induction of HO-1 by PAO and by heme may share some common pathways. oxophenylarsine 74-77 heme oxygenase 1 Homo sapiens 66-70 11768235-12 2001 Our results are consistent with the proposition that induction of HO-1 by PAO involves inhibition of specific PTP(s), and that the mechanisms of induction of HO-1 by PAO and by heme may share some common pathways. oxophenylarsine 166-169 heme oxygenase 1 Homo sapiens 158-162 11768235-12 2001 Our results are consistent with the proposition that induction of HO-1 by PAO involves inhibition of specific PTP(s), and that the mechanisms of induction of HO-1 by PAO and by heme may share some common pathways. Heme 177-181 heme oxygenase 1 Homo sapiens 66-70 11768235-12 2001 Our results are consistent with the proposition that induction of HO-1 by PAO involves inhibition of specific PTP(s), and that the mechanisms of induction of HO-1 by PAO and by heme may share some common pathways. Heme 177-181 heme oxygenase 1 Homo sapiens 158-162 11571246-3 2001 METHODS AND RESULTS: Infection of rat aortic smooth muscle cells with adenovirus carrying the human HO-1 gene (Adv-HO-1) resulted in a high-level expression of HO-1 protein, which effectively reduced the hemin-induced iron overload in these cells. Hemin 204-209 heme oxygenase 1 Homo sapiens 115-119 11571246-3 2001 METHODS AND RESULTS: Infection of rat aortic smooth muscle cells with adenovirus carrying the human HO-1 gene (Adv-HO-1) resulted in a high-level expression of HO-1 protein, which effectively reduced the hemin-induced iron overload in these cells. Iron 218-222 heme oxygenase 1 Homo sapiens 100-104 11571246-3 2001 METHODS AND RESULTS: Infection of rat aortic smooth muscle cells with adenovirus carrying the human HO-1 gene (Adv-HO-1) resulted in a high-level expression of HO-1 protein, which effectively reduced the hemin-induced iron overload in these cells. Iron 218-222 heme oxygenase 1 Homo sapiens 115-119 11572959-7 2001 Macrophage-inducible NOS generates NO to kill other cells, whereas HO1 generates bilirubin to exert antioxidant cytoprotective effects and also provides cytoprotection by facilitating iron extrusion from cells. Iron 184-188 heme oxygenase 1 Homo sapiens 67-70 11429414-3 2001 Once bound to its recognition DNA sequence termed antioxidant-responsive element or Maf-recognition element, Nrf2/small Maf induces a set of antioxidative stress genes, including heme oxygenase-1 and gamma-glutamylcysteine synthetase, whose products have been demonstrated to contribute to the scavenging of reactive oxygen species and to exhibit anti-inflammatory effects. Reactive Oxygen Species 308-331 heme oxygenase 1 Homo sapiens 179-195 11481283-4 2001 The degree of oxidative stress in cells was estimated by flow cytometric analysis of reactive oxygen intermediate (ROI)-induced 2",7"-dichlorodihydrofluorescein diacetate fluorescence and Northern blot analysis of heme oxygenase-1 (HO-1) mRNA induction. reactive oxygen intermediate 85-113 heme oxygenase 1 Homo sapiens 214-230 11481283-4 2001 The degree of oxidative stress in cells was estimated by flow cytometric analysis of reactive oxygen intermediate (ROI)-induced 2",7"-dichlorodihydrofluorescein diacetate fluorescence and Northern blot analysis of heme oxygenase-1 (HO-1) mRNA induction. reactive oxygen intermediate 85-113 heme oxygenase 1 Homo sapiens 232-236 11481283-7 2001 The degree of ROI production and HO-1 mRNA induction was greater in cells treated with 40% than 20% O(2), an effect that was also larger in old than young passaged cells. o(2) 100-104 heme oxygenase 1 Homo sapiens 33-37 11454666-9 2001 NOS inhibition by L-N(G)-nitroarginine methyl ester (L-NAME) or 1400 W abolished nitrite production and strongly reduced HO-1 expression. NG-Nitroarginine Methyl Ester 53-59 heme oxygenase 1 Homo sapiens 121-125 11448133-6 2001 In addition to HSP72, HSP32 was significantly increased by PGA1. prostaglandin A1 59-63 heme oxygenase 1 Homo sapiens 22-27 11448133-7 2001 The HSP32 induction was more vigorous with a marked increase with only 4 microg/ml of PGA1. prostaglandin A1 86-90 heme oxygenase 1 Homo sapiens 4-9 11551744-7 2001 Preconditioning induction of stress proteins (i.e., hemeoxygenase-1 and neuronal nitric oxide synthase) and hypothermia therapy suppress the generation of toxic reactive oxygen, lipid, and thiol species evoked by bioactive iron complexes in the brain. Sulfhydryl Compounds 189-194 heme oxygenase 1 Homo sapiens 52-67 11551744-7 2001 Preconditioning induction of stress proteins (i.e., hemeoxygenase-1 and neuronal nitric oxide synthase) and hypothermia therapy suppress the generation of toxic reactive oxygen, lipid, and thiol species evoked by bioactive iron complexes in the brain. Iron 223-227 heme oxygenase 1 Homo sapiens 52-67 11435909-3 2001 HO-1 metabolizes heme to the antioxidant bilirubin and carbon monoxide, and represents a powerful endogenous defensive mechanism against free radicals in many diseases. Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 11435909-3 2001 HO-1 metabolizes heme to the antioxidant bilirubin and carbon monoxide, and represents a powerful endogenous defensive mechanism against free radicals in many diseases. Bilirubin 41-50 heme oxygenase 1 Homo sapiens 0-4 11435909-3 2001 HO-1 metabolizes heme to the antioxidant bilirubin and carbon monoxide, and represents a powerful endogenous defensive mechanism against free radicals in many diseases. Carbon Monoxide 55-70 heme oxygenase 1 Homo sapiens 0-4 11435909-3 2001 HO-1 metabolizes heme to the antioxidant bilirubin and carbon monoxide, and represents a powerful endogenous defensive mechanism against free radicals in many diseases. Free Radicals 137-150 heme oxygenase 1 Homo sapiens 0-4 11435909-5 2001 In this study, we showed high expression of HO-1 in lesions of EAE, and demonstrated that hemin, an inducer of HO-1, inhibited EAE effectively. Hemin 90-95 heme oxygenase 1 Homo sapiens 44-48 11435909-5 2001 In this study, we showed high expression of HO-1 in lesions of EAE, and demonstrated that hemin, an inducer of HO-1, inhibited EAE effectively. Hemin 90-95 heme oxygenase 1 Homo sapiens 111-115 11435909-6 2001 In contrast, tin mesoporphyrin, an inhibitor of HO-1, markedly exacerbated EAE. tin mesoporphyrin 13-30 heme oxygenase 1 Homo sapiens 48-52 11454666-0 2001 Modulation of haem oxygenase-1 expression by nitric oxide and leukotrienes in zymosan-activated macrophages. Nitric Oxide 45-57 heme oxygenase 1 Homo sapiens 14-30 11454666-0 2001 Modulation of haem oxygenase-1 expression by nitric oxide and leukotrienes in zymosan-activated macrophages. Leukotrienes 62-74 heme oxygenase 1 Homo sapiens 14-30 11454666-0 2001 Modulation of haem oxygenase-1 expression by nitric oxide and leukotrienes in zymosan-activated macrophages. Zymosan 78-85 heme oxygenase 1 Homo sapiens 14-30 11274184-2 2001 Nrf2 regulates expression of genes encoding enzymes with antioxidant (e.g. heme oxygenase-1 (HO-1)) or xenobiotic detoxification (e.g. NAD(P)H:quinone oxidoreductase, glutathione S-transferase) functions via the stress- or antioxidant-response elements (StRE/ARE). nad(p)h 135-142 heme oxygenase 1 Homo sapiens 75-91 11454666-1 2001 Phagocytosis of unopsonized zymosan by RAW 264.7 macrophages upregulated protein expression of haem oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS) and cyclo-oxygenase-2 (COX-2) in a time- and concentration-dependent manner. Zymosan 28-35 heme oxygenase 1 Homo sapiens 95-111 11454666-1 2001 Phagocytosis of unopsonized zymosan by RAW 264.7 macrophages upregulated protein expression of haem oxygenase-1 (HO-1), inducible nitric oxide synthase (iNOS) and cyclo-oxygenase-2 (COX-2) in a time- and concentration-dependent manner. Zymosan 28-35 heme oxygenase 1 Homo sapiens 113-117 11454666-4 2001 The COX inhibitors indomethacin and NS398 weakly inhibited HO-1 expression but had no effect on iNOS and COX-2 expression or nitrite. Indomethacin 19-31 heme oxygenase 1 Homo sapiens 59-63 11454666-4 2001 The COX inhibitors indomethacin and NS398 weakly inhibited HO-1 expression but had no effect on iNOS and COX-2 expression or nitrite. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 36-41 heme oxygenase 1 Homo sapiens 59-63 11454666-5 2001 In contrast, the 5-lipoxygenase (5-LO) inhibitor ZM 230,487 significantly decreased HO-1, iNOS and nitrite, which were not affected by zileuton. zm 230 49-55 heme oxygenase 1 Homo sapiens 84-88 11454666-6 2001 Dexamethasone showed an inhibitory effect on HO-1 expression induced by zymosan. Dexamethasone 0-13 heme oxygenase 1 Homo sapiens 45-49 11454666-6 2001 Dexamethasone showed an inhibitory effect on HO-1 expression induced by zymosan. Zymosan 72-79 heme oxygenase 1 Homo sapiens 45-49 11454666-9 2001 NOS inhibition by L-N(G)-nitroarginine methyl ester (L-NAME) or 1400 W abolished nitrite production and strongly reduced HO-1 expression. l-n(g)-nitroarginine methyl ester 18-51 heme oxygenase 1 Homo sapiens 121-125 11274184-10 2001 These results indicate that ATF4 regulates basal and CdCl(2)-induced expression of the ho-1 gene in a cell-specific manner and possibly in a complex with Nrf2. Cadmium Chloride 53-60 heme oxygenase 1 Homo sapiens 87-91 11274184-2 2001 Nrf2 regulates expression of genes encoding enzymes with antioxidant (e.g. heme oxygenase-1 (HO-1)) or xenobiotic detoxification (e.g. NAD(P)H:quinone oxidoreductase, glutathione S-transferase) functions via the stress- or antioxidant-response elements (StRE/ARE). nad(p)h 135-142 heme oxygenase 1 Homo sapiens 93-97 23045067-1 2001 Heme oxygenase 1 and 2 activities are responsible for initiating most of the degradation of heme, although other enzyme pathways play a role as well. Heme 92-96 heme oxygenase 1 Homo sapiens 0-22 23045070-2 2001 This unit describes protocols for assessing the expression and regulation of specific HO-1 and HO-2 mRNAs by in situ hybridization using digoxigenin-tagged probes. Digoxigenin 137-148 heme oxygenase 1 Homo sapiens 86-90 11340095-1 2001 The possible beneficial effect of supplemental glutamine (Gln) in critically ill patients has been suggested to be mediated by the induction of the cytoprotective heat shock proteins (HSP)32 and HSP72. Glutamine 47-56 heme oxygenase 1 Homo sapiens 184-190 11340095-1 2001 The possible beneficial effect of supplemental glutamine (Gln) in critically ill patients has been suggested to be mediated by the induction of the cytoprotective heat shock proteins (HSP)32 and HSP72. Glutamine 58-61 heme oxygenase 1 Homo sapiens 184-190 11278277-5 2001 Expression of recombinant ATM in AT fibroblasts, however, reduced the extent of the effects of CdCl(2) on both c-Jun phosphorylation and heme oxygenase-1 induction. cdcl 95-99 heme oxygenase 1 Homo sapiens 137-153 11121422-0 2001 Disruption of an active site hydrogen bond converts human heme oxygenase-1 into a peroxidase. Hydrogen 29-37 heme oxygenase 1 Homo sapiens 58-74 11121422-1 2001 The crystal structure of heme oxygenase-1 suggests that Asp-140 may participate in a hydrogen bonding network involving ligands coordinated to the heme iron atom. Aspartic Acid 56-59 heme oxygenase 1 Homo sapiens 25-41 11121422-1 2001 The crystal structure of heme oxygenase-1 suggests that Asp-140 may participate in a hydrogen bonding network involving ligands coordinated to the heme iron atom. Hydrogen 85-93 heme oxygenase 1 Homo sapiens 25-41 11121422-1 2001 The crystal structure of heme oxygenase-1 suggests that Asp-140 may participate in a hydrogen bonding network involving ligands coordinated to the heme iron atom. Iron 152-156 heme oxygenase 1 Homo sapiens 25-41 11287117-0 2001 Heme oxygenase-1 induction by nitric oxide in RAW 264.7 macrophages is upregulated by a cyclo-oxygenase-2 inhibitor. Nitric Oxide 30-42 heme oxygenase 1 Homo sapiens 0-16 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. Nitric Oxide 110-122 heme oxygenase 1 Homo sapiens 166-170 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 134-150 heme oxygenase 1 Homo sapiens 54-70 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 134-150 heme oxygenase 1 Homo sapiens 72-76 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 134-150 heme oxygenase 1 Homo sapiens 166-170 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 152-156 heme oxygenase 1 Homo sapiens 54-70 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 152-156 heme oxygenase 1 Homo sapiens 72-76 11287117-1 2001 Unstimulated RAW 264.7 macrophages express negligible heme oxygenase-1 (HO-1) protein but incubation with the nitric oxide (NO) donor spermine nonoate (SPNO) induced HO-1 and weakly cyclo-oxygenase-2 (COX-2) protein. spermine nitric oxide complex 152-156 heme oxygenase 1 Homo sapiens 166-170 11287117-3 2001 Cells incubated with SPNO showed a strong increase in HO-1 mRNA levels after 4 h with a significant potentiation in the presence of SC58125, which did not modify HO-1 mRNA stability. spermine nitric oxide complex 21-25 heme oxygenase 1 Homo sapiens 54-58 11287117-3 2001 Cells incubated with SPNO showed a strong increase in HO-1 mRNA levels after 4 h with a significant potentiation in the presence of SC58125, which did not modify HO-1 mRNA stability. spermine nitric oxide complex 21-25 heme oxygenase 1 Homo sapiens 162-166 11688356-10 2001 In conclusion, carvedilol reduces oxidative stress and normalizes blood pressure; ecNOS remains upregulated while mRNA for transforming growth factor beta, a key fibrogenic cytokine, is reduced by carvedilol, which seems to preserve protective mechanisms such as NO and heme oxygenase-1 against long-term complications of oxidative stress, e.g., endothelial dysfunction, fibrogenesis and chronic rejection. Carvedilol 15-25 heme oxygenase 1 Homo sapiens 270-286 11688356-10 2001 In conclusion, carvedilol reduces oxidative stress and normalizes blood pressure; ecNOS remains upregulated while mRNA for transforming growth factor beta, a key fibrogenic cytokine, is reduced by carvedilol, which seems to preserve protective mechanisms such as NO and heme oxygenase-1 against long-term complications of oxidative stress, e.g., endothelial dysfunction, fibrogenesis and chronic rejection. Carvedilol 197-207 heme oxygenase 1 Homo sapiens 270-286 11247059-12 2001 CONCLUSIONS: The increase in bilirubin level by HO-1 might protect OSAHS patients from disorders related to hypoxemia. Bilirubin 29-38 heme oxygenase 1 Homo sapiens 48-52 11283857-4 2001 Administration of the NO donor, molsidomine (MOL) (3 mg. kg(-1)), during resuscitation attenuated the accumulation of HO-1 mRNA and protein and the rise in HO activity. Molsidomine 32-43 heme oxygenase 1 Homo sapiens 118-122 11283857-4 2001 Administration of the NO donor, molsidomine (MOL) (3 mg. kg(-1)), during resuscitation attenuated the accumulation of HO-1 mRNA and protein and the rise in HO activity. Molsidomine 45-48 heme oxygenase 1 Homo sapiens 118-122 11247059-0 2001 Increase in bilirubin levels of patients with obstructive sleep apnea in the morning--a possible explanation of induced heme oxygenase-1. Bilirubin 12-21 heme oxygenase 1 Homo sapiens 120-136 11171041-12 2001 In vitro studies with cultured keratinocytes revealed a potential role for reactive oxygen species (ROS), but not for growth factors and pro-inflammatory cytokines, as inducers of HO-1 expression in inflamed skin. Reactive Oxygen Species 75-98 heme oxygenase 1 Homo sapiens 180-184 11247059-1 2001 STUDY OBJECTIVES: In the absence of heme oxygenase-1 (HO-1), which catalyzes the oxidation of heme to generate carbon monoxide and indirect bilirubin, hypoxia induces severe right ventricular dilation and infarction. Heme 36-40 heme oxygenase 1 Homo sapiens 54-58 11247059-1 2001 STUDY OBJECTIVES: In the absence of heme oxygenase-1 (HO-1), which catalyzes the oxidation of heme to generate carbon monoxide and indirect bilirubin, hypoxia induces severe right ventricular dilation and infarction. Carbon Monoxide 111-126 heme oxygenase 1 Homo sapiens 36-52 11247059-1 2001 STUDY OBJECTIVES: In the absence of heme oxygenase-1 (HO-1), which catalyzes the oxidation of heme to generate carbon monoxide and indirect bilirubin, hypoxia induces severe right ventricular dilation and infarction. Carbon Monoxide 111-126 heme oxygenase 1 Homo sapiens 54-58 11247059-1 2001 STUDY OBJECTIVES: In the absence of heme oxygenase-1 (HO-1), which catalyzes the oxidation of heme to generate carbon monoxide and indirect bilirubin, hypoxia induces severe right ventricular dilation and infarction. Bilirubin 140-149 heme oxygenase 1 Homo sapiens 36-52 11247059-1 2001 STUDY OBJECTIVES: In the absence of heme oxygenase-1 (HO-1), which catalyzes the oxidation of heme to generate carbon monoxide and indirect bilirubin, hypoxia induces severe right ventricular dilation and infarction. Bilirubin 140-149 heme oxygenase 1 Homo sapiens 54-58 11245628-3 2001 There are three isoforms of HO: HO-1 is inducible by inflammatory cytokines and oxidants, including nitric oxide (NO), whereas HO-2 and HO-3 are expressed constitutively. Nitric Oxide 100-112 heme oxygenase 1 Homo sapiens 32-36 11171043-0 2001 Tobacco-smoke-inducible human haem oxygenase-1 gene expression: role of distinct transcription factors and reactive oxygen intermediates. reactive oxygen 107-122 heme oxygenase 1 Homo sapiens 30-46 11171043-1 2001 Exposure of eukaryotic cells to a variety of reactive-oxygen-intermediate (ROI)-mediated sources of cellular injury, including heavy metals and UV radiation, induces the expression of heat-shock (HS) and stress-related genes among which is a 32-34 kDa protein identified as inducible haem oxygenase-1 (HO-1). reactive 45-53 heme oxygenase 1 Homo sapiens 284-300 11171043-1 2001 Exposure of eukaryotic cells to a variety of reactive-oxygen-intermediate (ROI)-mediated sources of cellular injury, including heavy metals and UV radiation, induces the expression of heat-shock (HS) and stress-related genes among which is a 32-34 kDa protein identified as inducible haem oxygenase-1 (HO-1). reactive 45-53 heme oxygenase 1 Homo sapiens 302-306 11171043-1 2001 Exposure of eukaryotic cells to a variety of reactive-oxygen-intermediate (ROI)-mediated sources of cellular injury, including heavy metals and UV radiation, induces the expression of heat-shock (HS) and stress-related genes among which is a 32-34 kDa protein identified as inducible haem oxygenase-1 (HO-1). Oxygen 54-60 heme oxygenase 1 Homo sapiens 284-300 11171043-1 2001 Exposure of eukaryotic cells to a variety of reactive-oxygen-intermediate (ROI)-mediated sources of cellular injury, including heavy metals and UV radiation, induces the expression of heat-shock (HS) and stress-related genes among which is a 32-34 kDa protein identified as inducible haem oxygenase-1 (HO-1). Oxygen 54-60 heme oxygenase 1 Homo sapiens 302-306 11171043-8 2001 We report an inhibition of NF-kappaB activation by TS, no effect on AP-1 and a strong activation of CdRE-binding activity, whereas cadmium chelation from TS only partially prevented HO-1 induction. Cadmium 131-138 heme oxygenase 1 Homo sapiens 182-186 11171043-9 2001 H(2)O(2) also activated the CdRE-binding activity, and pretreatment with N-acetyl-L-cysteine, which replenishes the intracellular levels of GSH, suppressed, in TS-treated cells, both the CdRE-binding activity and the increased HO-1 expression. Hydrogen Peroxide 0-8 heme oxygenase 1 Homo sapiens 227-231 11171043-9 2001 H(2)O(2) also activated the CdRE-binding activity, and pretreatment with N-acetyl-L-cysteine, which replenishes the intracellular levels of GSH, suppressed, in TS-treated cells, both the CdRE-binding activity and the increased HO-1 expression. Acetylcysteine 73-92 heme oxygenase 1 Homo sapiens 227-231 11171043-9 2001 H(2)O(2) also activated the CdRE-binding activity, and pretreatment with N-acetyl-L-cysteine, which replenishes the intracellular levels of GSH, suppressed, in TS-treated cells, both the CdRE-binding activity and the increased HO-1 expression. Glutathione 140-143 heme oxygenase 1 Homo sapiens 227-231 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Heme 57-61 heme oxygenase 1 Homo sapiens 0-16 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Heme 57-61 heme oxygenase 1 Homo sapiens 18-22 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 0-16 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Carbon Monoxide 88-103 heme oxygenase 1 Homo sapiens 18-22 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Carbon Monoxide 105-107 heme oxygenase 1 Homo sapiens 0-16 11950143-2 2001 Heme oxygenase isozymes, HO-1, HO-2 and HO-3, are HSP32 protein cognates, with a known function of catalyzing the isomer specific oxidation of the heme molecule, including that of NO synthase. Heme 147-151 heme oxygenase 1 Homo sapiens 25-44 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Carbon Monoxide 105-107 heme oxygenase 1 Homo sapiens 18-22 11950143-2 2001 Heme oxygenase isozymes, HO-1, HO-2 and HO-3, are HSP32 protein cognates, with a known function of catalyzing the isomer specific oxidation of the heme molecule, including that of NO synthase. Heme 147-151 heme oxygenase 1 Homo sapiens 50-55 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Biliverdine 113-123 heme oxygenase 1 Homo sapiens 0-16 11950143-11 2001 In the case of severe tissue injury, such as compression injury, HO-1 is induced and colocalizes with cGMP and pro-apoptotic oncogenes. Cyclic GMP 102-106 heme oxygenase 1 Homo sapiens 65-69 11194943-4 2001 Heme oxygenase-1 (HO-1) is an inducible enzyme degrading heme into the gaseous mediator carbon monoxide (CO) and biliverdin, a local antioxidant. Biliverdine 113-123 heme oxygenase 1 Homo sapiens 18-22 11135063-10 2001 CONCLUSION: We conclude that up-regulation of HO-1 occurs in the kidney in humans and rats repetitively exposed to heme proteins. Heme 115-119 heme oxygenase 1 Homo sapiens 46-50 11124818-1 2001 This study was designed to determine changes in expression of heme oxygenase (HO)-1, the stress-inducible and carbon monoxide-producing enzyme, in normotensive and portal hypertensive human livers. Carbon Monoxide 110-125 heme oxygenase 1 Homo sapiens 62-83 11208917-3 2001 We have also shown that reactive iron (Fe3+) and cGMP staining spatially resemble that of HO-1; which, in turn, colocalizes in motor neurons with transcription factors: Fas-associated protein containing death domain (FADD), tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p53. Iron 33-37 heme oxygenase 1 Homo sapiens 90-94 11208917-3 2001 We have also shown that reactive iron (Fe3+) and cGMP staining spatially resemble that of HO-1; which, in turn, colocalizes in motor neurons with transcription factors: Fas-associated protein containing death domain (FADD), tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p53. ferric sulfate 39-43 heme oxygenase 1 Homo sapiens 90-94 11208917-3 2001 We have also shown that reactive iron (Fe3+) and cGMP staining spatially resemble that of HO-1; which, in turn, colocalizes in motor neurons with transcription factors: Fas-associated protein containing death domain (FADD), tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) and p53. Cyclic GMP 49-53 heme oxygenase 1 Homo sapiens 90-94 11208917-11 2001 Tissue Fe3+ and cGMP staining were increased and prominently mapped below the site of injury, where cGMP colocalized with HO-1 in the nucleus of the motor neurons. ferric sulfate 7-11 heme oxygenase 1 Homo sapiens 122-126 11208917-11 2001 Tissue Fe3+ and cGMP staining were increased and prominently mapped below the site of injury, where cGMP colocalized with HO-1 in the nucleus of the motor neurons. Cyclic GMP 16-20 heme oxygenase 1 Homo sapiens 122-126 11208917-11 2001 Tissue Fe3+ and cGMP staining were increased and prominently mapped below the site of injury, where cGMP colocalized with HO-1 in the nucleus of the motor neurons. Cyclic GMP 100-104 heme oxygenase 1 Homo sapiens 122-126 11018038-5 2000 TGF-beta1 treatment in conjunction with actinomycin D or cycloheximide demonstrated that induction of HO-1 mRNA requires de novo transcription and, in part, protein synthesis. Dactinomycin 40-53 heme oxygenase 1 Homo sapiens 102-106 11018038-5 2000 TGF-beta1 treatment in conjunction with actinomycin D or cycloheximide demonstrated that induction of HO-1 mRNA requires de novo transcription and, in part, protein synthesis. Cycloheximide 57-70 heme oxygenase 1 Homo sapiens 102-106 10942763-0 2000 Replacement of the distal glycine 139 transforms human heme oxygenase-1 into a peroxidase. Glycine 26-33 heme oxygenase 1 Homo sapiens 55-71 11078701-6 2000 15-d-PGJ(2) and its precursors, but not the thiazolidinediones, induced gene expression for heme oxygenase-1 (HO-1), a stress-related protein, and strongly inhibited interleukin-1 (IL-1)-induced nitric oxide (NO) production. 15-d-pgj 0-8 heme oxygenase 1 Homo sapiens 92-108 11078701-6 2000 15-d-PGJ(2) and its precursors, but not the thiazolidinediones, induced gene expression for heme oxygenase-1 (HO-1), a stress-related protein, and strongly inhibited interleukin-1 (IL-1)-induced nitric oxide (NO) production. 15-d-pgj 0-8 heme oxygenase 1 Homo sapiens 110-114 11078701-6 2000 15-d-PGJ(2) and its precursors, but not the thiazolidinediones, induced gene expression for heme oxygenase-1 (HO-1), a stress-related protein, and strongly inhibited interleukin-1 (IL-1)-induced nitric oxide (NO) production. Nitric Oxide 195-207 heme oxygenase 1 Homo sapiens 92-108 11078701-6 2000 15-d-PGJ(2) and its precursors, but not the thiazolidinediones, induced gene expression for heme oxygenase-1 (HO-1), a stress-related protein, and strongly inhibited interleukin-1 (IL-1)-induced nitric oxide (NO) production. Nitric Oxide 195-207 heme oxygenase 1 Homo sapiens 110-114 11078701-8 2000 On the other hand, sodium arsenite, a known activator of the stress response pathway, induced HO-1 mRNA expression but lacked a promoting effect on TG production. sodium arsenite 19-34 heme oxygenase 1 Homo sapiens 94-98 11076874-0 2000 Inhibition of inducible nitric oxide synthase in the human intestinal epithelial cell line, DLD-1, by the inducers of heme oxygenase 1, bismuth salts, heme, and nitric oxide donors. nitric 24-30 heme oxygenase 1 Homo sapiens 118-134 11076874-0 2000 Inhibition of inducible nitric oxide synthase in the human intestinal epithelial cell line, DLD-1, by the inducers of heme oxygenase 1, bismuth salts, heme, and nitric oxide donors. Nitric Oxide 24-36 heme oxygenase 1 Homo sapiens 118-134 11076874-8 2000 This inhibitory effect was abolished by the HO-1 specific inhibitor tin protoporphyrin. tin protoporphyrin IX 68-86 heme oxygenase 1 Homo sapiens 44-48 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Heme 63-67 heme oxygenase 1 Homo sapiens 81-85 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Iron 134-138 heme oxygenase 1 Homo sapiens 63-79 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Iron 134-138 heme oxygenase 1 Homo sapiens 81-85 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 63-79 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Biliverdine 140-150 heme oxygenase 1 Homo sapiens 81-85 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Carbon Monoxide 156-171 heme oxygenase 1 Homo sapiens 63-79 11085891-2 2000 Among the consequences of oxidative stress is the induction of heme oxygenase-1 (HO-1), an inducible isozyme that metabolizes heme to iron, biliverdin, and carbon monoxide. Carbon Monoxide 156-171 heme oxygenase 1 Homo sapiens 81-85 11115065-0 2000 Dopamine induces the expression of heme oxygenase-1 by human endothelial cells in vitro. Dopamine 0-8 heme oxygenase 1 Homo sapiens 35-51 11115065-2 2000 To study the mechanisms underlying the organ protection associated with the administration of DA prior to transplantation, we questioned whether DA induces the antioxidative enzyme heme oxygenase-1 (HO-1) in cultured endothelial cells. Dopamine 145-147 heme oxygenase 1 Homo sapiens 199-203 11115065-5 2000 RESULTS: The presence of DA resulted in a dose- and time-dependent up-regulation of HO-1 both on RNA and protein level, whereas HO-1 was barely detectable under basal conditions. Dopamine 25-27 heme oxygenase 1 Homo sapiens 84-88 11115065-8 2000 The addition of the antioxidant agents ascorbic acid and N-acetyl-cysteine both lead to dose-dependent inhibition of DA-mediated HO-1 induction. Ascorbic Acid 39-52 heme oxygenase 1 Homo sapiens 129-133 11115065-8 2000 The addition of the antioxidant agents ascorbic acid and N-acetyl-cysteine both lead to dose-dependent inhibition of DA-mediated HO-1 induction. Acetylcysteine 57-74 heme oxygenase 1 Homo sapiens 129-133 11115065-10 2000 CONCLUSION: We conclude that DA induces the expression of the protective enzyme HO-1 in cultured endothelial cells by an oxidative mechanism. Dopamine 29-31 heme oxygenase 1 Homo sapiens 80-84 10942763-1 2000 The human heme oxygenase-1 crystal structure suggests that Gly-139 and Gly-143 interact directly with iron-bound ligands. Glycine 59-62 heme oxygenase 1 Homo sapiens 10-26 10942763-1 2000 The human heme oxygenase-1 crystal structure suggests that Gly-139 and Gly-143 interact directly with iron-bound ligands. Glycine 71-74 heme oxygenase 1 Homo sapiens 10-26 10942763-1 2000 The human heme oxygenase-1 crystal structure suggests that Gly-139 and Gly-143 interact directly with iron-bound ligands. Iron 102-106 heme oxygenase 1 Homo sapiens 10-26 11032845-0 2000 Nitric oxide-inducible expression of heme oxygenase-1 in human cells. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 37-53 11032845-3 2000 Nitric oxide (NO) induces HO-1 in many cell types, but the specific contribution of transcriptional or post-transcriptional effects to this induction have remained unresolved. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 26-30 11032845-5 2000 We used a specific NO scavenger (2-(4-carboxylphenyl)-4,4,5,5-tetramethylimidazolin-1-oxyl 3-oxide) that completely prevented the inducible expression of HO-1 by NO, pointing to direct signaling action of NO in this induction. 2-(4-carboxylphenyl)-4,4,5,5-tetramethylimidazolin-1-oxyl 3-oxide 33-98 heme oxygenase 1 Homo sapiens 154-158 11007950-1 2000 Heme oxygenase (HO)-1 is the inducible isoform of the rate-limiting enzyme of heme degradation. Heme 78-82 heme oxygenase 1 Homo sapiens 0-21 11015442-5 2000 Exposure of ECs to exogenous CO, under inhibition of HO-1 activity by SnPPIX, substitutes HO-1 in preventing EC apoptosis. Carbon Monoxide 29-31 heme oxygenase 1 Homo sapiens 53-57 11015442-0 2000 Carbon monoxide generated by heme oxygenase 1 suppresses endothelial cell apoptosis. Carbon Monoxide 0-15 heme oxygenase 1 Homo sapiens 29-45 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Heme 14-18 heme oxygenase 1 Homo sapiens 119-141 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Heme 14-18 heme oxygenase 1 Homo sapiens 143-149 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Carbon Monoxide 198-213 heme oxygenase 1 Homo sapiens 119-141 11015442-1 2000 Heme oxygenase 1 (HO-1) inhibits apoptosis by regulating cellular prooxidant iron. Iron 77-81 heme oxygenase 1 Homo sapiens 0-22 11015442-5 2000 Exposure of ECs to exogenous CO, under inhibition of HO-1 activity by SnPPIX, substitutes HO-1 in preventing EC apoptosis. Carbon Monoxide 29-31 heme oxygenase 1 Homo sapiens 90-94 11015442-2 2000 We now show that there is an additional mechanism by which HO-1 inhibits apoptosis, namely by generating the gaseous molecule carbon monoxide (CO). Carbon Monoxide 126-141 heme oxygenase 1 Homo sapiens 59-63 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Carbon Monoxide 198-213 heme oxygenase 1 Homo sapiens 143-149 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Carbon Monoxide 215-217 heme oxygenase 1 Homo sapiens 119-141 11015442-2 2000 We now show that there is an additional mechanism by which HO-1 inhibits apoptosis, namely by generating the gaseous molecule carbon monoxide (CO). Carbon Monoxide 143-145 heme oxygenase 1 Homo sapiens 59-63 11015442-5 2000 Exposure of ECs to exogenous CO, under inhibition of HO-1 activity by SnPPIX, substitutes HO-1 in preventing EC apoptosis. tin protoporphyrin IX 70-76 heme oxygenase 1 Homo sapiens 53-57 11015442-3 2000 Overexpression of HO-1, or induction of HO-1 expression by heme, protects endothelial cells (ECs) from apoptosis. Heme 59-63 heme oxygenase 1 Homo sapiens 40-44 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Carbon Monoxide 215-217 heme oxygenase 1 Homo sapiens 143-149 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Iron 220-224 heme oxygenase 1 Homo sapiens 119-141 11015442-4 2000 When HO-1 enzymatic activity is blocked by tin protoporphyrin (SnPPIX) or the action of CO is inhibited by hemoglobin (Hb), HO-1 no longer prevents EC apoptosis while these reagents do not affect the antiapoptotic action of bcl-2. tin protoporphyrin IX 43-61 heme oxygenase 1 Homo sapiens 5-9 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Iron 220-224 heme oxygenase 1 Homo sapiens 143-149 11015442-8 2000 Specific inhibition of p38 MAPK activation by the pyridinyl imidazol SB203580 or through overexpression of a p38 MAPK dominant negative mutant abrogated the antiapoptotic effect of HO-1. pyridinyl imidazol 50-68 heme oxygenase 1 Homo sapiens 181-185 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Iron 226-228 heme oxygenase 1 Homo sapiens 119-141 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Iron 226-228 heme oxygenase 1 Homo sapiens 143-149 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Biliverdine 234-244 heme oxygenase 1 Homo sapiens 119-141 10985695-1 2000 Extracellular heme derived from hemoglobin following hemorrhage or released from dying cells induces the expression of heme oxygenase-1 (HO-1, HSP-32) which metabolizes heme to the gaseous mediator carbon monoxide (CO), iron (Fe) and biliverdin. Biliverdine 234-244 heme oxygenase 1 Homo sapiens 143-149 11015442-4 2000 When HO-1 enzymatic activity is blocked by tin protoporphyrin (SnPPIX) or the action of CO is inhibited by hemoglobin (Hb), HO-1 no longer prevents EC apoptosis while these reagents do not affect the antiapoptotic action of bcl-2. tin protoporphyrin IX 63-69 heme oxygenase 1 Homo sapiens 5-9 11015442-8 2000 Specific inhibition of p38 MAPK activation by the pyridinyl imidazol SB203580 or through overexpression of a p38 MAPK dominant negative mutant abrogated the antiapoptotic effect of HO-1. SB 203580 69-77 heme oxygenase 1 Homo sapiens 181-185 11023535-8 2000 Our results suggest that increased sequestration of iron as a consequence of induced HO-1 might be involved in the adaptive protection after HBO treatment and that the induction of DNA damage is not the trigger for adaptive protection. Iron 52-56 heme oxygenase 1 Homo sapiens 85-89 10997923-0 2000 Mechanisms underlying induction of heme oxygenase-1 by nitric oxide in renal tubular epithelial cells. Nitric Oxide 55-67 heme oxygenase 1 Homo sapiens 35-51 10997923-1 2000 We examined whether nitric oxide-generating agents influence expression of heme oxygenase-1 (HO-1) in renal proximal tubular epithelial cells, LLC-PK(1) cells, and the mechanisms underlying any such effects. Nitric Oxide 20-32 heme oxygenase 1 Homo sapiens 75-91 10997923-1 2000 We examined whether nitric oxide-generating agents influence expression of heme oxygenase-1 (HO-1) in renal proximal tubular epithelial cells, LLC-PK(1) cells, and the mechanisms underlying any such effects. Nitric Oxide 20-32 heme oxygenase 1 Homo sapiens 93-97 10997923-2 2000 In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. Nitric Oxide 25-37 heme oxygenase 1 Homo sapiens 73-77 10997923-2 2000 In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. Nitroprusside 44-64 heme oxygenase 1 Homo sapiens 73-77 10997923-2 2000 In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. Nitric Oxide 191-203 heme oxygenase 1 Homo sapiens 73-77 10997923-2 2000 In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. Nitric Oxide 191-203 heme oxygenase 1 Homo sapiens 73-77 10997923-2 2000 In sublytic amounts, the nitric oxide donor sodium nitroprusside induced HO-1 mRNA and protein and HO activity in a dose-dependent and time-dependent fashion; this induction was specific for nitric oxide since the nitric oxide scavenger carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide significantly reduced such induction. carboxy-2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide 237-299 heme oxygenase 1 Homo sapiens 73-77 10997923-8 2000 We thus provide the novel description of the induction of HO-1 in renal proximal tubular epithelial cells exposed to nitric oxide donors and provide the first demonstration in kidney-derived cells for the involvement of a redox-based mechanism in such expression. Nitric Oxide 117-129 heme oxygenase 1 Homo sapiens 58-62 10997923-9 2000 We also demonstrate that, in LLC-PK(1) cells exposed to nitric oxide donors, chelatable iron is involved in eliciting the HO-1 response observed at lower concentrations of these donors, and in mediating the cytotoxic effects of these donors when present in higher concentrations. Nitric Oxide 56-68 heme oxygenase 1 Homo sapiens 122-126 10997923-9 2000 We also demonstrate that, in LLC-PK(1) cells exposed to nitric oxide donors, chelatable iron is involved in eliciting the HO-1 response observed at lower concentrations of these donors, and in mediating the cytotoxic effects of these donors when present in higher concentrations. Iron 88-92 heme oxygenase 1 Homo sapiens 122-126 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Heme 64-68 heme oxygenase 1 Homo sapiens 0-16 10874044-0 2000 Mechanism of heme oxygenase-1 gene activation by cadmium in MCF-7 mammary epithelial cells. Cadmium 49-56 heme oxygenase 1 Homo sapiens 13-29 10874044-5 2000 Exposure of MCF-7 cells to 10 micrometer CdCl(2) stimulates phosphorylation of ERK, JNK, and p38 mitogen-activated protein kinases, implicating one or more of these signaling pathways in ho-1 gene induction. cdcl 41-45 heme oxygenase 1 Homo sapiens 187-191 10874044-6 2000 SB203580, an inhibitor of p38, diminishes cadmium-stimulated pE1-luc expression and HO-1 mRNA levels by up to 70-80%. SB 203580 0-8 heme oxygenase 1 Homo sapiens 84-88 10874044-13 2000 Taken together, these results indicate that cadmium induces ho-1 gene expression via sequential activation of the p38 kinase pathway and Nrf2. Cadmium 44-51 heme oxygenase 1 Homo sapiens 60-64 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Heme 64-68 heme oxygenase 1 Homo sapiens 18-22 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Biliverdine 72-82 heme oxygenase 1 Homo sapiens 0-16 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Biliverdine 72-82 heme oxygenase 1 Homo sapiens 18-22 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Iron 89-93 heme oxygenase 1 Homo sapiens 0-16 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Iron 89-93 heme oxygenase 1 Homo sapiens 18-22 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 0-16 11053673-2 2000 Heme oxygenase-1 (HO-1) is a 32kDa stress protein that degrades heme to biliverdin, free iron and carbon monoxide. Carbon Monoxide 98-113 heme oxygenase 1 Homo sapiens 18-22 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. Iron 88-92 heme oxygenase 1 Homo sapiens 63-67 10975414-6 2000 Total RNA from EPS was used for gene expression of heme oxygenase-1 (HO-1) and granzyme B. eps 15-18 heme oxygenase 1 Homo sapiens 51-67 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. Iron 88-92 heme oxygenase 1 Homo sapiens 122-126 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. tin mesoporphyrin 139-156 heme oxygenase 1 Homo sapiens 63-67 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. tin mesoporphyrin 139-156 heme oxygenase 1 Homo sapiens 122-126 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. Dexamethasone 161-174 heme oxygenase 1 Homo sapiens 63-67 11053673-4 2000 In these cells and in rat astroglia transfected with the human HO-1 gene, mitochondrial iron trapping is abrogated by the HO-1 inhibitors, tin-mesoporphyrin and dexamethasone. Dexamethasone 161-174 heme oxygenase 1 Homo sapiens 122-126 11053673-7 2000 Collectively, our findings suggest that HO-1 over-expression contributes to the pathological iron deposition and mitochondrial damage documented in these aging-related neurodegenerative disorders. Iron 93-97 heme oxygenase 1 Homo sapiens 40-44 10906077-6 2000 Treatments of isolated tubules with either sodium arsenite, known to induce HO-1, or hematin, an HO substrate, resulted in 4.4- and 1.8-fold, respectively, increases in cGMP levels. sodium arsenite 43-58 heme oxygenase 1 Homo sapiens 76-80 10906077-6 2000 Treatments of isolated tubules with either sodium arsenite, known to induce HO-1, or hematin, an HO substrate, resulted in 4.4- and 1.8-fold, respectively, increases in cGMP levels. Cyclic GMP 169-173 heme oxygenase 1 Homo sapiens 76-80 10906077-9 2000 These findings, demonstrating for the first time a link between HO-1 activity in Sertoli cells and sGC-dependent cGMP production in seminiferous tubules, suggest a functional role of CO in the human testis. Cyclic GMP 113-117 heme oxygenase 1 Homo sapiens 64-68 10894889-2 2000 Arsenite is also a potent inducer of heme oxygenase (HO)-1. arsenite 0-8 heme oxygenase 1 Homo sapiens 37-58 10788478-2 2000 The stress protein heme oxygenase-1 (HO-1) is induced in endothelial cells exposed to nitric oxide (NO)-releasing agents, and this process is finely modulated by thiols (Foresti, R., Clark, J. E., Green, C. J., and Motterlini R. (1997) J. Biol. Nitric Oxide 86-98 heme oxygenase 1 Homo sapiens 19-35 11229523-12 2000 In addition, surface redox-active copper is the first link shown for the concomitant regulation of HO-1 and MT-1 and is required for the activation of the amino-terminal c-Jun kinase (JNK) by heme-hemopexin. Copper 34-40 heme oxygenase 1 Homo sapiens 99-112 10938455-6 2000 In lymphocytes isolated from individuals receiving antioxidant supplements and subjected to a heat shock in the presence of the free radical generator 2, 2"-azobis-(2-amidinopropane)-dihydrochloride, enhanced synthesis of heat shock proteins hsp 105, hsp 90, hsp 70, and hsp 40 by contrast with decreased synthesis of heme oxygenase HO-1 (hsp 32) were noted. Free Radicals 128-140 heme oxygenase 1 Homo sapiens 333-337 10938455-6 2000 In lymphocytes isolated from individuals receiving antioxidant supplements and subjected to a heat shock in the presence of the free radical generator 2, 2"-azobis-(2-amidinopropane)-dihydrochloride, enhanced synthesis of heat shock proteins hsp 105, hsp 90, hsp 70, and hsp 40 by contrast with decreased synthesis of heme oxygenase HO-1 (hsp 32) were noted. Free Radicals 128-140 heme oxygenase 1 Homo sapiens 339-345 10938455-6 2000 In lymphocytes isolated from individuals receiving antioxidant supplements and subjected to a heat shock in the presence of the free radical generator 2, 2"-azobis-(2-amidinopropane)-dihydrochloride, enhanced synthesis of heat shock proteins hsp 105, hsp 90, hsp 70, and hsp 40 by contrast with decreased synthesis of heme oxygenase HO-1 (hsp 32) were noted. 2,2'-azobis(2-amidinopropane) 151-198 heme oxygenase 1 Homo sapiens 333-337 10938455-6 2000 In lymphocytes isolated from individuals receiving antioxidant supplements and subjected to a heat shock in the presence of the free radical generator 2, 2"-azobis-(2-amidinopropane)-dihydrochloride, enhanced synthesis of heat shock proteins hsp 105, hsp 90, hsp 70, and hsp 40 by contrast with decreased synthesis of heme oxygenase HO-1 (hsp 32) were noted. 2,2'-azobis(2-amidinopropane) 151-198 heme oxygenase 1 Homo sapiens 339-345 10814519-2 2000 Unexpectedly, here we show that expression of HO-1 mRNA is repressed by hypoxia in primary cultures of human umbilical vein endothelial cells (HUVECs), but is increased by cobalt chloride (CoCl(2)) that is known to mimic hypoxia. cobaltous chloride 172-187 heme oxygenase 1 Homo sapiens 46-50 10814519-2 2000 Unexpectedly, here we show that expression of HO-1 mRNA is repressed by hypoxia in primary cultures of human umbilical vein endothelial cells (HUVECs), but is increased by cobalt chloride (CoCl(2)) that is known to mimic hypoxia. cobaltous chloride 189-196 heme oxygenase 1 Homo sapiens 46-50 10814519-4 2000 Therefore, hypoxia and cobalt showed opposing effects on HO-1 mRNA expression, despite activation of hypoxia-inducible factor 1. Cobalt 23-29 heme oxygenase 1 Homo sapiens 57-61 10814519-5 2000 The half-life of HO-1 mRNA was not changed by hypoxia, but was significantly prolonged by CoCl(2). cobaltous chloride 90-97 heme oxygenase 1 Homo sapiens 17-21 10839994-2 2000 In several circumstances, this cytoprotective effect has been attributed to increased generation of the antioxidant bilirubin during haem degradation by HO-1. Bilirubin 116-125 heme oxygenase 1 Homo sapiens 153-157 10839994-3 2000 However, a direct implication for HO-1-derived bilirubin in protection against oxidative stress remains to be established. Bilirubin 47-56 heme oxygenase 1 Homo sapiens 34-38 10839994-5 2000 We found that hemin-mediated increase in HO-1 protein expression and haem oxygenase activity is associated with augmented bilirubin levels. Bilirubin 122-131 heme oxygenase 1 Homo sapiens 41-45 10839994-8 2000 Interestingly, this protective effect was manifest only when cells were actively producing bilirubin as a consequence of increased haem availability and utilization by HO-1. Bilirubin 91-100 heme oxygenase 1 Homo sapiens 168-172 10839994-11 2000 Our findings provide direct evidence that bilirubin generated after up-regulation of the HO-1 pathway is cytoprotective against oxidative stress. Bilirubin 42-51 heme oxygenase 1 Homo sapiens 89-93 10788478-2 2000 The stress protein heme oxygenase-1 (HO-1) is induced in endothelial cells exposed to nitric oxide (NO)-releasing agents, and this process is finely modulated by thiols (Foresti, R., Clark, J. E., Green, C. J., and Motterlini R. (1997) J. Biol. Nitric Oxide 86-98 heme oxygenase 1 Homo sapiens 37-41 10788478-2 2000 The stress protein heme oxygenase-1 (HO-1) is induced in endothelial cells exposed to nitric oxide (NO)-releasing agents, and this process is finely modulated by thiols (Foresti, R., Clark, J. E., Green, C. J., and Motterlini R. (1997) J. Biol. Sulfhydryl Compounds 162-168 heme oxygenase 1 Homo sapiens 19-35 10788478-2 2000 The stress protein heme oxygenase-1 (HO-1) is induced in endothelial cells exposed to nitric oxide (NO)-releasing agents, and this process is finely modulated by thiols (Foresti, R., Clark, J. E., Green, C. J., and Motterlini R. (1997) J. Biol. Sulfhydryl Compounds 162-168 heme oxygenase 1 Homo sapiens 37-41 10788478-8 2000 A series of antioxidant agents did not prevent the elevation in heme oxygenase activity by hypoxia; however, the precursor of glutathione synthesis and thiol donor, N-acetylcysteine, completely abolished HO-1 induction. Glutathione 126-137 heme oxygenase 1 Homo sapiens 204-208 10788478-8 2000 A series of antioxidant agents did not prevent the elevation in heme oxygenase activity by hypoxia; however, the precursor of glutathione synthesis and thiol donor, N-acetylcysteine, completely abolished HO-1 induction. Sulfhydryl Compounds 152-157 heme oxygenase 1 Homo sapiens 204-208 10788478-8 2000 A series of antioxidant agents did not prevent the elevation in heme oxygenase activity by hypoxia; however, the precursor of glutathione synthesis and thiol donor, N-acetylcysteine, completely abolished HO-1 induction. Acetylcysteine 165-181 heme oxygenase 1 Homo sapiens 204-208 10807584-6 2000 In vitro studies in human renal proximal tubule cells demonstrate that hemin, an inducer of HO-1, significantly attenuated cisplatin-induced apoptosis and necrosis, whereas inhibition of HO-1 enzyme activity reversed the cytoprotective effect. Hemin 71-76 heme oxygenase 1 Homo sapiens 92-96 10788478-10 2000 These results indicate that intracellular interaction of thiols with NO is an important determinant in the mechanism leading to HO-1 induction by reduced oxygen levels. Sulfhydryl Compounds 57-63 heme oxygenase 1 Homo sapiens 128-132 10807584-6 2000 In vitro studies in human renal proximal tubule cells demonstrate that hemin, an inducer of HO-1, significantly attenuated cisplatin-induced apoptosis and necrosis, whereas inhibition of HO-1 enzyme activity reversed the cytoprotective effect. Cisplatin 123-132 heme oxygenase 1 Homo sapiens 92-96 10788478-10 2000 These results indicate that intracellular interaction of thiols with NO is an important determinant in the mechanism leading to HO-1 induction by reduced oxygen levels. Oxygen 154-160 heme oxygenase 1 Homo sapiens 128-132 10788478-11 2000 We suggest that in addition to oxidative stress, HO-1 gene expression can be regulated by redox reactions involving NO and S-nitrosothiols (nitrosative stress), emphasizing a versatile role for the heme oxygenase pathway in the cellular adaptation to a variety of stressful conditions. S-Nitrosothiols 123-138 heme oxygenase 1 Homo sapiens 49-53 10780998-5 2000 Incubation of cells with NO donors, spermine nonoate (SPNO) and S-nitroso-N-acetylpenicillamine (SNAP), induced a dose- and time-dependent expression of HO-1 protein. spermine nitric oxide complex 36-52 heme oxygenase 1 Homo sapiens 153-157 10807735-0 2000 Heme oxygenase-1 is a cGMP-inducible endothelial protein and mediates the cytoprotective action of nitric oxide. Cyclic GMP 22-26 heme oxygenase 1 Homo sapiens 0-16 10807735-0 2000 Heme oxygenase-1 is a cGMP-inducible endothelial protein and mediates the cytoprotective action of nitric oxide. Nitric Oxide 99-111 heme oxygenase 1 Homo sapiens 0-16 10807735-2 2000 By use of Western blotting, cell viability analysis, and antisense technique, the present study investigates the involvement of HO-1 in endothelial protection induced by the clinically used nitric oxide (NO) donor molsidomine (specifically, its active metabolite 3-morpholinosydnonimine [SIN-1]) and the second messenger cGMP. Nitric Oxide 190-202 heme oxygenase 1 Homo sapiens 128-132 10807735-2 2000 By use of Western blotting, cell viability analysis, and antisense technique, the present study investigates the involvement of HO-1 in endothelial protection induced by the clinically used nitric oxide (NO) donor molsidomine (specifically, its active metabolite 3-morpholinosydnonimine [SIN-1]) and the second messenger cGMP. Molsidomine 214-225 heme oxygenase 1 Homo sapiens 128-132 10807735-4 2000 HO-1 induction by SIN-1 was inhibited in the presence of the NO scavenger phenyl-4,4,5,5,-tetramethylimidazoline-1-oxyl-3-oxide and the soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazole[4, 3-a]quinoxalin-1-one. phenyl-4,4,5,5,-tetramethylimidazoline-1-oxyl-3-oxide 74-127 heme oxygenase 1 Homo sapiens 0-4 10780998-0 2000 Enhanced expression of haem oxygenase-1 by nitric oxide and antiinflammatory drugs in NIH 3T3 fibroblasts. Nitric Oxide 43-55 heme oxygenase 1 Homo sapiens 23-39 10807735-4 2000 HO-1 induction by SIN-1 was inhibited in the presence of the NO scavenger phenyl-4,4,5,5,-tetramethylimidazoline-1-oxyl-3-oxide and the soluble guanylyl cyclase inhibitor 1H-[1,2,4]oxadiazole[4, 3-a]quinoxalin-1-one. 1h-[1,2,4]oxadiazole[4, 3-a]quinoxalin-1-one 171-215 heme oxygenase 1 Homo sapiens 0-4 10807735-8 2000 Pretreatment of cells with a phosphorothioate-linked HO-1 antisense oligonucleotide prevented protection by SIN-1 or 8-bromo-cGMP against tumor necrosis factor-alpha cytotoxicity, whereas sense and scrambled HO-1 were without effect under these conditions. Parathion 29-45 heme oxygenase 1 Homo sapiens 53-57 10807735-8 2000 Pretreatment of cells with a phosphorothioate-linked HO-1 antisense oligonucleotide prevented protection by SIN-1 or 8-bromo-cGMP against tumor necrosis factor-alpha cytotoxicity, whereas sense and scrambled HO-1 were without effect under these conditions. Parathion 29-45 heme oxygenase 1 Homo sapiens 208-212 10807735-8 2000 Pretreatment of cells with a phosphorothioate-linked HO-1 antisense oligonucleotide prevented protection by SIN-1 or 8-bromo-cGMP against tumor necrosis factor-alpha cytotoxicity, whereas sense and scrambled HO-1 were without effect under these conditions. Oligonucleotides 68-83 heme oxygenase 1 Homo sapiens 53-57 10807735-8 2000 Pretreatment of cells with a phosphorothioate-linked HO-1 antisense oligonucleotide prevented protection by SIN-1 or 8-bromo-cGMP against tumor necrosis factor-alpha cytotoxicity, whereas sense and scrambled HO-1 were without effect under these conditions. Oligonucleotides 68-83 heme oxygenase 1 Homo sapiens 208-212 10807735-9 2000 Our results show for the first time that HO-1 is a cGMP-sensitive endothelial gene and establish conclusively a causal relationship between HO-1 induction and endothelial protection by the NO/cGMP system. Cyclic GMP 51-55 heme oxygenase 1 Homo sapiens 41-45 10807735-9 2000 Our results show for the first time that HO-1 is a cGMP-sensitive endothelial gene and establish conclusively a causal relationship between HO-1 induction and endothelial protection by the NO/cGMP system. Cyclic GMP 51-55 heme oxygenase 1 Homo sapiens 140-144 10807735-9 2000 Our results show for the first time that HO-1 is a cGMP-sensitive endothelial gene and establish conclusively a causal relationship between HO-1 induction and endothelial protection by the NO/cGMP system. Cyclic GMP 192-196 heme oxygenase 1 Homo sapiens 41-45 10807735-9 2000 Our results show for the first time that HO-1 is a cGMP-sensitive endothelial gene and establish conclusively a causal relationship between HO-1 induction and endothelial protection by the NO/cGMP system. Cyclic GMP 192-196 heme oxygenase 1 Homo sapiens 140-144 10780998-5 2000 Incubation of cells with NO donors, spermine nonoate (SPNO) and S-nitroso-N-acetylpenicillamine (SNAP), induced a dose- and time-dependent expression of HO-1 protein. spermine nitric oxide complex 54-58 heme oxygenase 1 Homo sapiens 153-157 10780998-5 2000 Incubation of cells with NO donors, spermine nonoate (SPNO) and S-nitroso-N-acetylpenicillamine (SNAP), induced a dose- and time-dependent expression of HO-1 protein. S-Nitroso-N-Acetylpenicillamine 64-95 heme oxygenase 1 Homo sapiens 153-157 10780998-5 2000 Incubation of cells with NO donors, spermine nonoate (SPNO) and S-nitroso-N-acetylpenicillamine (SNAP), induced a dose- and time-dependent expression of HO-1 protein. S-Nitroso-N-Acetylpenicillamine 97-101 heme oxygenase 1 Homo sapiens 153-157 10780998-8 2000 COX-2 protein was weakly induced by SPNO in basal conditions and in the presence of LPS a synergy for HO-1 and COX-2 protein expression was observed. spermine nitric oxide complex 36-40 heme oxygenase 1 Homo sapiens 102-106 10780998-10 2000 Our results indicate that reactive oxygen species participate in the inductive effect of NO donors or LPS on HO-1 expression, whereas endogenous NO production may play a role in the mechanism of the synergy exhibited by SPNO and LPS on HO-1 and COX-2 expression. Reactive Oxygen Species 26-49 heme oxygenase 1 Homo sapiens 109-113 10780998-10 2000 Our results indicate that reactive oxygen species participate in the inductive effect of NO donors or LPS on HO-1 expression, whereas endogenous NO production may play a role in the mechanism of the synergy exhibited by SPNO and LPS on HO-1 and COX-2 expression. spermine nitric oxide complex 220-224 heme oxygenase 1 Homo sapiens 236-240 10780998-13 2000 The selective COX-2 inhibitors SC58125 and NS398 as well as the non-selective COX inhibitor, indomethacin, strongly reduced PGE(2) synthesis and showed a synergy with NO donors in HO-1 and COX-2 induction. 1-((4-methylsulfonyl)phenyl)-3-trifluoromethyl-5-(4-fluorophenyl)pyrazole 31-38 heme oxygenase 1 Homo sapiens 180-184 10872747-5 2000 Pre-treatment of the cells with PD98059, a selective ERK pathway inhibitor, and SB203580, a p38 MAPK inhibitor, blocked the induction of HO-1 by the NO donor in a dose-dependent manner. 2-(2-amino-3-methoxyphenyl)-4H-1-benzopyran-4-one 32-39 heme oxygenase 1 Homo sapiens 137-141 10872745-5 2000 We had previously shown that inhibitors of HO-1 and the mitochondrial permeability transition pore (MTP) block the uptake of mitochondrial iron in astrocytes exposed to the pro-oxidant effects of dopamine and several pro-inflammatory cytokines. Iron 139-143 heme oxygenase 1 Homo sapiens 43-47 10872745-5 2000 We had previously shown that inhibitors of HO-1 and the mitochondrial permeability transition pore (MTP) block the uptake of mitochondrial iron in astrocytes exposed to the pro-oxidant effects of dopamine and several pro-inflammatory cytokines. Dopamine 196-204 heme oxygenase 1 Homo sapiens 43-47 10872747-5 2000 Pre-treatment of the cells with PD98059, a selective ERK pathway inhibitor, and SB203580, a p38 MAPK inhibitor, blocked the induction of HO-1 by the NO donor in a dose-dependent manner. SB 203580 80-88 heme oxygenase 1 Homo sapiens 137-141 10780998-13 2000 The selective COX-2 inhibitors SC58125 and NS398 as well as the non-selective COX inhibitor, indomethacin, strongly reduced PGE(2) synthesis and showed a synergy with NO donors in HO-1 and COX-2 induction. N-(2-cyclohexyloxy-4-nitrophenyl)methanesulfonamide 43-48 heme oxygenase 1 Homo sapiens 180-184 10872747-12 2000 ERK and p38 inhibitors also suppressed induction of HO-1 by SNAP and GSNO. snap 60-64 heme oxygenase 1 Homo sapiens 52-56 10872745-7 2000 Thus, the marked enhancement of HO-1 expression previously demonstrated in AD-affected neurons and astroglia may transduce amyloid (oxidative) stress into the abnormal patterns of iron deposition and mitochondrial insufficiency characteristic of this disease. Iron 180-184 heme oxygenase 1 Homo sapiens 32-36 10872745-8 2000 Finally, in experiments employing cytotoxic concentrations of A40, we provide evidence that inhibition of HO-1 transcription and related mitochondrial iron deposition may be an important mechanism by which DEX protects tissues subjected to amyloid stress. Dexamethasone 206-209 heme oxygenase 1 Homo sapiens 106-110 10780998-13 2000 The selective COX-2 inhibitors SC58125 and NS398 as well as the non-selective COX inhibitor, indomethacin, strongly reduced PGE(2) synthesis and showed a synergy with NO donors in HO-1 and COX-2 induction. Indomethacin 93-105 heme oxygenase 1 Homo sapiens 180-184 10872747-0 2000 Nitric oxide induces heme oxygenase-1 via mitogen-activated protein kinases ERK and p38. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 21-37 10872747-12 2000 ERK and p38 inhibitors also suppressed induction of HO-1 by SNAP and GSNO. S-Nitrosoglutathione 69-73 heme oxygenase 1 Homo sapiens 52-56 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Bilirubin 264-273 heme oxygenase 1 Homo sapiens 16-20 10872747-13 2000 The increase in HO-1 mRNA was inhibited by actinomycin D and cycloheximide, but not by NAC, and was not mimicked by the lipophilic cGMP analogue, 8-bromo-cGMP, suggesting that NO-mediated induction required de novo RNA and protein synthesis and was unrelated to cGMP and redox signaling. Dactinomycin 43-56 heme oxygenase 1 Homo sapiens 16-20 10872747-13 2000 The increase in HO-1 mRNA was inhibited by actinomycin D and cycloheximide, but not by NAC, and was not mimicked by the lipophilic cGMP analogue, 8-bromo-cGMP, suggesting that NO-mediated induction required de novo RNA and protein synthesis and was unrelated to cGMP and redox signaling. Cycloheximide 61-74 heme oxygenase 1 Homo sapiens 16-20 10872747-13 2000 The increase in HO-1 mRNA was inhibited by actinomycin D and cycloheximide, but not by NAC, and was not mimicked by the lipophilic cGMP analogue, 8-bromo-cGMP, suggesting that NO-mediated induction required de novo RNA and protein synthesis and was unrelated to cGMP and redox signaling. Cyclic GMP 154-158 heme oxygenase 1 Homo sapiens 16-20 10872748-7 2000 Analysis of MT-1, MT-2 and HO-1 expression, through Western and Northern blotting, indicates that there is a variable pattern of induction and suppression of these two genes following hypoxia preconditioning alone as well as after kainic acid-induced seizures compared to non-preconditioned animals. Kainic Acid 231-242 heme oxygenase 1 Homo sapiens 27-31 10773020-7 2000 In addition, we investigated the possibility that the human HO-1 gene altered the expression of the endogenous rat enzyme after administration of cobalt chloride s.c. Cobalt chloride administration resulted in increased HO activity in all tissues examined in rats transduced with the human HO-1 gene to the same degree as in nontransduced rats. cobaltous chloride 146-161 heme oxygenase 1 Homo sapiens 60-64 10773020-7 2000 In addition, we investigated the possibility that the human HO-1 gene altered the expression of the endogenous rat enzyme after administration of cobalt chloride s.c. Cobalt chloride administration resulted in increased HO activity in all tissues examined in rats transduced with the human HO-1 gene to the same degree as in nontransduced rats. cobaltous chloride 167-182 heme oxygenase 1 Homo sapiens 60-64 10773020-7 2000 In addition, we investigated the possibility that the human HO-1 gene altered the expression of the endogenous rat enzyme after administration of cobalt chloride s.c. Cobalt chloride administration resulted in increased HO activity in all tissues examined in rats transduced with the human HO-1 gene to the same degree as in nontransduced rats. cobaltous chloride 167-182 heme oxygenase 1 Homo sapiens 290-294 10773020-9 2000 The increase in HO activity after adenoviral-mediated human HO-1 transfer was associated with a decrease in microsomal heme-CYP and CYP activity. Heme 119-123 heme oxygenase 1 Homo sapiens 60-64 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Carbon Monoxide 244-259 heme oxygenase 1 Homo sapiens 16-20 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Carbon Monoxide 244-259 heme oxygenase 1 Homo sapiens 62-66 10872747-2 2000 Nitric oxide (NO) is among the extensive array of stimuli that induce HO-1. Nitric Oxide 0-12 heme oxygenase 1 Homo sapiens 70-74 10872747-4 2000 In the present study, we have demonstrated that exposure of HeLa cells to the NO donor, sodium nitroprusside (SNP), results in concentration and time-dependent increase in HO-1 mRNA and activation of MAPKs: ERK (ERK1 and ERK2) and p38 pathways, but not SAPK/JNK pathway. Nitroprusside 88-108 heme oxygenase 1 Homo sapiens 172-176 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Carbon Monoxide 244-259 heme oxygenase 1 Homo sapiens 62-66 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Bilirubin 264-273 heme oxygenase 1 Homo sapiens 62-66 10773020-10 2000 The increase in HO-1 activity after adenovirus-mediated human HO-1 gene transfer may prove useful as a means of selectively increasing enzyme activity in a specific organ and regulating homeostasis by modulation of vasoactive molecules such as carbon monoxide and bilirubin and, in addition, providing a means of delivering the human HO-1 gene for experimental purposes. Bilirubin 264-273 heme oxygenase 1 Homo sapiens 62-66 10766788-7 2000 Based on sequence identity with the mammalian enzymes the proximal ligand in HO-1 (His-25) and HO-2 (His-45) is conserved (His-20) in the bacterial enzyme. Histidine 83-86 heme oxygenase 1 Homo sapiens 77-81 10766788-7 2000 Based on sequence identity with the mammalian enzymes the proximal ligand in HO-1 (His-25) and HO-2 (His-45) is conserved (His-20) in the bacterial enzyme. Histidine 101-104 heme oxygenase 1 Homo sapiens 77-81 10766788-7 2000 Based on sequence identity with the mammalian enzymes the proximal ligand in HO-1 (His-25) and HO-2 (His-45) is conserved (His-20) in the bacterial enzyme. Histidine 101-104 heme oxygenase 1 Homo sapiens 77-81 10746601-1 2000 BACKGROUND: Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that catalyzes the degradation of heme to biliverdin. Heme 97-101 heme oxygenase 1 Homo sapiens 12-28 10733947-1 2000 Heme oxygenase-1 is the heme catabolic enzyme induced in human dermal fibroblasts by environmental stress. Heme 24-28 heme oxygenase 1 Homo sapiens 0-16 10733947-4 2000 The relationship between heme oxygenase-1 activation and iron metabolism was investigated by measurement of activities of both cytosolic and mitochondrial cis-aconitase enzymes. Iron 57-61 heme oxygenase 1 Homo sapiens 25-41 10733947-6 2000 We propose that modulation of cis-aconitase activity at the translational level by an increase of cellular iron is an important consequence of heme oxygenase-1 activation. Iron 107-111 heme oxygenase 1 Homo sapiens 143-159 11232594-2 2000 The precipitating cause of such oxidative stress may be misregulated iron homeostasis because there are profound alterations in heme oxygenase-1 (HO-1), redox-active iron, and iron regulatory proteins. Iron 69-73 heme oxygenase 1 Homo sapiens 128-144 11232594-2 2000 The precipitating cause of such oxidative stress may be misregulated iron homeostasis because there are profound alterations in heme oxygenase-1 (HO-1), redox-active iron, and iron regulatory proteins. Iron 69-73 heme oxygenase 1 Homo sapiens 146-150 10746601-1 2000 BACKGROUND: Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that catalyzes the degradation of heme to biliverdin. Heme 97-101 heme oxygenase 1 Homo sapiens 30-34 10746601-1 2000 BACKGROUND: Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that catalyzes the degradation of heme to biliverdin. Biliverdine 105-115 heme oxygenase 1 Homo sapiens 12-28 10746601-1 2000 BACKGROUND: Heme oxygenase-1 (HO-1) is a 32-kDa stress protein that catalyzes the degradation of heme to biliverdin. Biliverdine 105-115 heme oxygenase 1 Homo sapiens 30-34 10746601-7 2000 The AACD group exhibited plasma HO-1 concentrations (1.06 +/- 0.33 microg/mL) intermediate between, but not different from, those of the AD patients and NEC. aacd 4-8 heme oxygenase 1 Homo sapiens 32-36 10631150-3 2000 A (GT)n dinucleotide repeat in the 5"-flanking region of human HO-1 gene shows length polymorphism and could modulate the level of gene transcription. n dinucleotide 6-20 heme oxygenase 1 Homo sapiens 63-67 10681502-0 2000 Reaction intermediates and single turnover rate constants for the oxidation of heme by human heme oxygenase-1. Heme 79-83 heme oxygenase 1 Homo sapiens 93-109 10681514-4 2000 In transfected cells overexpressing HO-1, the activity of heme oxygenase was increased, and conversely, the level of tau protein was dramatically decreased when compared with antisense HO-1 or CEP transfected cells. Heme 58-62 heme oxygenase 1 Homo sapiens 36-40 10653390-9 2000 To explore the hypothesis that these peptides exert their immunosuppressive effect by altering HO-1 activity, animals were treated with iron protoporphyrin, an inducer of HO-1 activity, or tin protoporphyrin, an inhibitor of HO-1. Heme 136-155 heme oxygenase 1 Homo sapiens 171-175 10653390-9 2000 To explore the hypothesis that these peptides exert their immunosuppressive effect by altering HO-1 activity, animals were treated with iron protoporphyrin, an inducer of HO-1 activity, or tin protoporphyrin, an inhibitor of HO-1. Heme 136-155 heme oxygenase 1 Homo sapiens 171-175 10733587-7 2000 Activation of the heme oxygenase 1 promoter by arsenite or cadmium and activation of the cadmium-inducible metallothionein promoter also were unaffected by the expression of mutant SWI-SNF components. Cadmium 59-66 heme oxygenase 1 Homo sapiens 18-34 10672026-3 2000 We report here that the known antioxidant pyrrolidinedithiocarbamate (PDTC) leads to time and dose dependent activation of heat-shock protein 70 (Hsp70) as well as Hsp32 in EC. pyrrolidine dithiocarbamic acid 42-68 heme oxygenase 1 Homo sapiens 164-169 10672026-3 2000 We report here that the known antioxidant pyrrolidinedithiocarbamate (PDTC) leads to time and dose dependent activation of heat-shock protein 70 (Hsp70) as well as Hsp32 in EC. pyrrolidine dithiocarbamic acid 70-74 heme oxygenase 1 Homo sapiens 164-169 10631150-8 2000 H2O2 exposure up-regulated the transcriptional activity of the HO-1 promoter/luciferase fusion genes with (GT)16 or (GT)20 but did not do so with (GT)29 or (GT)38. Hydrogen Peroxide 0-4 heme oxygenase 1 Homo sapiens 63-67 10631150-9 2000 These findings suggest that the large size of a (GT)n repeat in the HO-1 gene promoter may reduce HO-1 inducibility by reactive oxygen species in cigarette smoke, thereby resulting in the development of CPE. Reactive Oxygen Species 119-142 heme oxygenase 1 Homo sapiens 68-72 10644516-0 2000 Induction of heme oxygenase-1 by hypoxia and free radicals in human dermal fibroblasts. Free Radicals 45-58 heme oxygenase 1 Homo sapiens 13-29 10644516-1 2000 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme catabolism and presumably is involved in cellular iron homeostasis. Iron 115-119 heme oxygenase 1 Homo sapiens 0-16 10644516-1 2000 Heme oxygenase-1 (HO-1) catalyzes the rate-limiting step in heme catabolism and presumably is involved in cellular iron homeostasis. Iron 115-119 heme oxygenase 1 Homo sapiens 18-22 10863530-8 2000 Mammalian cells also sense and respond to sub-toxic levels of nitric oxide, activating expression of heme oxygenase 1 through stabilization of its mRNA. Nitric Oxide 62-74 heme oxygenase 1 Homo sapiens 101-117 10644516-6 2000 In contrast, HO-1 induction by the oxidants, H(2)O(2) and carbonyl cyanide m-chlorophenylhydrazone (CCCP) was significantly attenuated in the presence of free radical scavengers. Hydrogen Peroxide 45-53 heme oxygenase 1 Homo sapiens 13-17 10644516-6 2000 In contrast, HO-1 induction by the oxidants, H(2)O(2) and carbonyl cyanide m-chlorophenylhydrazone (CCCP) was significantly attenuated in the presence of free radical scavengers. Carbonyl Cyanide m-Chlorophenyl Hydrazone 58-98 heme oxygenase 1 Homo sapiens 13-17 10644516-6 2000 In contrast, HO-1 induction by the oxidants, H(2)O(2) and carbonyl cyanide m-chlorophenylhydrazone (CCCP) was significantly attenuated in the presence of free radical scavengers. Carbonyl Cyanide m-Chlorophenyl Hydrazone 100-104 heme oxygenase 1 Homo sapiens 13-17 10644516-8 2000 Iron depletion by desferrioxamine mesylate, a specific iron complexon, completely inhibited hypoxic stimulation of HO-1 but did not attenuate the effect of H(2)O(2) and CCCP on HO-1 mRNA. Iron 0-4 heme oxygenase 1 Homo sapiens 115-119 10644516-8 2000 Iron depletion by desferrioxamine mesylate, a specific iron complexon, completely inhibited hypoxic stimulation of HO-1 but did not attenuate the effect of H(2)O(2) and CCCP on HO-1 mRNA. Deferoxamine 18-42 heme oxygenase 1 Homo sapiens 115-119 10644516-8 2000 Iron depletion by desferrioxamine mesylate, a specific iron complexon, completely inhibited hypoxic stimulation of HO-1 but did not attenuate the effect of H(2)O(2) and CCCP on HO-1 mRNA. Iron 55-59 heme oxygenase 1 Homo sapiens 115-119 10644516-9 2000 Addition of Fe(2+), Fe(3+), or holo-transferrin to fibroblasts increased levels of HO-1 mRNA. ammonium ferrous sulfate 12-18 heme oxygenase 1 Homo sapiens 83-87 10644516-9 2000 Addition of Fe(2+), Fe(3+), or holo-transferrin to fibroblasts increased levels of HO-1 mRNA. ferric sulfate 20-26 heme oxygenase 1 Homo sapiens 83-87 15693275-6 2000 Our results show that Homocysteine (Hcy) and oxidized LDL (oxLDL) enhance the activity and expression of oxidative stress markers, such as NFkB and heme oxygenase 1. Homocysteine 22-34 heme oxygenase 1 Homo sapiens 148-164 15693275-6 2000 Our results show that Homocysteine (Hcy) and oxidized LDL (oxLDL) enhance the activity and expression of oxidative stress markers, such as NFkB and heme oxygenase 1. Homocysteine 36-39 heme oxygenase 1 Homo sapiens 148-164 10631150-9 2000 These findings suggest that the large size of a (GT)n repeat in the HO-1 gene promoter may reduce HO-1 inducibility by reactive oxygen species in cigarette smoke, thereby resulting in the development of CPE. Reactive Oxygen Species 119-142 heme oxygenase 1 Homo sapiens 98-102 10842752-4 2000 In fact, in oxidative stress in vitro, HO-1 is protective (91-94) but within a narrow threshold of overexpression (93,94) in some models, since iron released in the HO reaction may obviate any cytoprotective effect (Fig. Iron 144-148 heme oxygenase 1 Homo sapiens 39-43 10564189-2 1999 Our results show that especially Zn(2+) and Cd(2+) are inducers of 70-kDa (HSP70), 60-kDa (HSP60), 32-kDa (HSP32), and 27-kDa (HSP27) HSPs. Zinc 33-35 heme oxygenase 1 Homo sapiens 107-112 10627295-8 2000 HO-1 immunostaining was low in the placenta but intense on the CTB within the placental bed. ctb 63-66 heme oxygenase 1 Homo sapiens 0-4 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Biliverdine 83-93 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Biliverdine 83-93 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 95-110 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 95-110 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 112-114 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 112-114 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 121-125 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 121-125 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 127-129 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 127-129 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 165-167 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Carbon Monoxide 165-167 heme oxygenase 1 Homo sapiens 60-64 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 172-174 heme oxygenase 1 Homo sapiens 42-58 10589693-4 1999 Induction of the heme catabolizing enzyme heme oxygenase-1 (HO-1), which generates biliverdin, carbon monoxide (CO), and iron (Fe), may provide such a mechanism, as CO and Fe are two negative modulators of iNOS activity and expression. Iron 172-174 heme oxygenase 1 Homo sapiens 60-64 10589693-6 1999 Glomerular HO-1 expression in nephritic animals was upregulated by treatment with hemin (30 micromol/kg body wt). Hemin 82-87 heme oxygenase 1 Homo sapiens 11-15 10589693-12 1999 Hemin treatment of nephritic animals resulted in upregulation of glomerular HO-1 levels and a two- to threefold reduction in glomerular iNOS mRNA levels. Hemin 0-5 heme oxygenase 1 Homo sapiens 76-80 10589693-15 1999 These studies demonstrate that in glomerular immune injury, hemin treatment upregulates glomerular HO-1 with an attendant downregulation of iNOS expression, and thus points to regulatory interaction between the two systems. Hemin 60-65 heme oxygenase 1 Homo sapiens 99-103 10629191-4 2000 The product of the hemO gene is homologous to enzymes that degrade heme; 21% of its amino acid residues are identical, and 44% are similar, to those of the human heme oxygenase-1. Heme 67-71 heme oxygenase 1 Homo sapiens 162-178 10630670-3 1999 However, as attempts are made to unravel the mechanisms by which HO-1 is induced and as we discover that CO, iron and bilirubin may be important effector molecules, we are learning to appreciate that heme oxygenases may be central to the regulation of many physiological and pathophysiological processes besides their established function in heme catabolism. Heme 200-204 heme oxygenase 1 Homo sapiens 65-69 10581397-0 1999 Regulation of heme oxygenase-1 expression by dopamine in cultured C6 glioma and primary astrocytes. Dopamine 45-53 heme oxygenase 1 Homo sapiens 14-30 10581397-1 1999 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in heme catabolism, tissue iron homeostasis and the cellular response to oxidative stress. Iron 83-87 heme oxygenase 1 Homo sapiens 0-16 10581397-1 1999 Heme oxygenase-1 (HO-1) is an inducible enzyme involved in heme catabolism, tissue iron homeostasis and the cellular response to oxidative stress. Iron 83-87 heme oxygenase 1 Homo sapiens 18-22 10581397-2 1999 Elevated HO-1 expression in astrocytes has been observed in association with abnormal iron deposition and increased oxidative stress in Parkinson"s disease (PD). Iron 86-90 heme oxygenase 1 Homo sapiens 9-13 10581397-4 1999 Here we report that dopamine is a potent inducer of HO-1. Dopamine 20-28 heme oxygenase 1 Homo sapiens 52-56 10581397-6 1999 When the time-course of HO-1 expression was compared between dopamine and hemin, the latter induced the gene immediately while the former did so with a lag. Dopamine 61-69 heme oxygenase 1 Homo sapiens 24-28 10581397-6 1999 When the time-course of HO-1 expression was compared between dopamine and hemin, the latter induced the gene immediately while the former did so with a lag. Hemin 74-79 heme oxygenase 1 Homo sapiens 24-28 10581397-8 1999 However, cycloheximide blocked both hemin- and dopamine-induced HO-1 expression, suggesting that both pathways may involve proteins with short half-lives. Cycloheximide 9-22 heme oxygenase 1 Homo sapiens 64-68 10581397-8 1999 However, cycloheximide blocked both hemin- and dopamine-induced HO-1 expression, suggesting that both pathways may involve proteins with short half-lives. Dopamine 47-55 heme oxygenase 1 Homo sapiens 64-68 10581397-9 1999 Ascorbic acid blocked dopamine induction of HO-1 but had no effect on hemin-induced expression. Ascorbic Acid 0-13 heme oxygenase 1 Homo sapiens 44-48 10581397-9 1999 Ascorbic acid blocked dopamine induction of HO-1 but had no effect on hemin-induced expression. Dopamine 22-30 heme oxygenase 1 Homo sapiens 44-48 10581397-12 1999 These results suggest that dopamine, released or secreted from affected neurons, may trigger HO-1 expression in neighboring astrocytes. Dopamine 27-35 heme oxygenase 1 Homo sapiens 93-97 10581397-13 1999 HO-1 and its metabolites could then contribute to the oxidative stress and iron deposition associated with PD. Iron 75-79 heme oxygenase 1 Homo sapiens 0-4 10564189-2 1999 Our results show that especially Zn(2+) and Cd(2+) are inducers of 70-kDa (HSP70), 60-kDa (HSP60), 32-kDa (HSP32), and 27-kDa (HSP27) HSPs. Cadmium 44-46 heme oxygenase 1 Homo sapiens 107-112 10490932-4 1999 Additionally, we studied HO-1 expression in experimental models of adhesion molecule expression produced by heme in endothelial cells, and the relationship of HO-1 expression to the induced adhesion molecules. Heme 108-112 heme oxygenase 1 Homo sapiens 25-29 10547244-1 1999 OBJECTIVES: Oxidative stress and inflammation induce the expression of heme oxygenase-1, which produces carbon monoxide (CO), and nitric oxide synthase, which produces nitric oxide (NO). Carbon Monoxide 104-119 heme oxygenase 1 Homo sapiens 71-87 10547244-1 1999 OBJECTIVES: Oxidative stress and inflammation induce the expression of heme oxygenase-1, which produces carbon monoxide (CO), and nitric oxide synthase, which produces nitric oxide (NO). Carbon Monoxide 121-123 heme oxygenase 1 Homo sapiens 71-87 10564544-2 1999 We have shown that the inflammatory cytokines, interleukin-6 (IL-6) and heme-induced HO-1 gene expression, were suppressed by dexamethasone (Dex) in a sustained manner. Dexamethasone 126-139 heme oxygenase 1 Homo sapiens 85-89 10564544-2 1999 We have shown that the inflammatory cytokines, interleukin-6 (IL-6) and heme-induced HO-1 gene expression, were suppressed by dexamethasone (Dex) in a sustained manner. Dexamethasone 141-144 heme oxygenase 1 Homo sapiens 85-89 10564544-4 1999 Endothelial cells treated with heme (10 microM) and IL-6 (25 ng/ml), increased HO-1 mRNA 15- and 60-fold, respectively. Heme 31-35 heme oxygenase 1 Homo sapiens 79-83 10564544-7 1999 Several human HO-1 promoter-drive chloramphenicol acetyltransferase (CAT) constructs were examined to analyze IL-6 and Dex-mediated modulation of the HO-1 gene in endothelial cells. Dexamethasone 119-122 heme oxygenase 1 Homo sapiens 150-154 10517538-2 1999 The expression of several genes (including heme oxygenase-1, HO-1; collagenase; the CL100 phosphatase and the nuclear oncogenes, c-fos and c-jun) is induced following physiological doses of UVA to cells and this effect can be strongly enhanced by removing intracellular glutathione or enhancing singlet oxygen lifetime. Glutathione 270-281 heme oxygenase 1 Homo sapiens 43-59 10517538-2 1999 The expression of several genes (including heme oxygenase-1, HO-1; collagenase; the CL100 phosphatase and the nuclear oncogenes, c-fos and c-jun) is induced following physiological doses of UVA to cells and this effect can be strongly enhanced by removing intracellular glutathione or enhancing singlet oxygen lifetime. Glutathione 270-281 heme oxygenase 1 Homo sapiens 61-65 10517538-2 1999 The expression of several genes (including heme oxygenase-1, HO-1; collagenase; the CL100 phosphatase and the nuclear oncogenes, c-fos and c-jun) is induced following physiological doses of UVA to cells and this effect can be strongly enhanced by removing intracellular glutathione or enhancing singlet oxygen lifetime. Singlet Oxygen 295-309 heme oxygenase 1 Homo sapiens 43-59 10517538-2 1999 The expression of several genes (including heme oxygenase-1, HO-1; collagenase; the CL100 phosphatase and the nuclear oncogenes, c-fos and c-jun) is induced following physiological doses of UVA to cells and this effect can be strongly enhanced by removing intracellular glutathione or enhancing singlet oxygen lifetime. Singlet Oxygen 295-309 heme oxygenase 1 Homo sapiens 61-65 10517538-3 1999 We have observed that heme is released from microsomal heme-containing proteins by UVA and other oxidants and that activation of HO-1 expression by UVA correlates with levels of heme release. Heme 22-26 heme oxygenase 1 Homo sapiens 129-133 10517538-4 1999 UVA radiation also leads to an increase in labile iron pools (either directly or via HO-1) and eventual increases in ferritin levels. Iron 50-54 heme oxygenase 1 Homo sapiens 85-89 10490932-17 1999 It is possible that amelioration of the heme-induced oxidative stress and expression of ICAM-I is due, in part, to the induction of HO-1 activity. Heme 40-44 heme oxygenase 1 Homo sapiens 132-136 10490932-9 1999 Endothelial cells exposed to heme elicited increased HO activity, which was prevented (70%) by HO-1 antisense ODNs. Heme 29-33 heme oxygenase 1 Homo sapiens 95-99 10490932-11 1999 Addition of HO-1 antisense ODNs prevented heme degradation and resulted in elevation of microsomal heme. Heme 42-46 heme oxygenase 1 Homo sapiens 12-16 10490932-11 1999 Addition of HO-1 antisense ODNs prevented heme degradation and resulted in elevation of microsomal heme. Heme 99-103 heme oxygenase 1 Homo sapiens 12-16 10490624-6 1999 5"-AMP and to a lesser extent 5"-ADP inhibit c-Src when combined with either HSP-27 or HSP-32. Adenosine Monophosphate 0-6 heme oxygenase 1 Homo sapiens 87-93 10490932-13 1999 Incubation of endothelial cells with HO-1 antisense enhanced heme-dependent increase of ICAM-1. Heme 61-65 heme oxygenase 1 Homo sapiens 37-41 10491455-3 1999 Exhaled carbon monoxide (CO), a product of heme degradation by heme oxygenase 1 (HO-1) which is induced by inflammatory cytokines and oxidants, was therefore tested as a non-invasive marker of airway inflammation and oxidative stress. Carbon Monoxide 8-23 heme oxygenase 1 Homo sapiens 63-79 10491455-3 1999 Exhaled carbon monoxide (CO), a product of heme degradation by heme oxygenase 1 (HO-1) which is induced by inflammatory cytokines and oxidants, was therefore tested as a non-invasive marker of airway inflammation and oxidative stress. Carbon Monoxide 8-23 heme oxygenase 1 Homo sapiens 81-85 10491455-3 1999 Exhaled carbon monoxide (CO), a product of heme degradation by heme oxygenase 1 (HO-1) which is induced by inflammatory cytokines and oxidants, was therefore tested as a non-invasive marker of airway inflammation and oxidative stress. Carbon Monoxide 25-27 heme oxygenase 1 Homo sapiens 63-79 10491455-3 1999 Exhaled carbon monoxide (CO), a product of heme degradation by heme oxygenase 1 (HO-1) which is induced by inflammatory cytokines and oxidants, was therefore tested as a non-invasive marker of airway inflammation and oxidative stress. Carbon Monoxide 25-27 heme oxygenase 1 Homo sapiens 81-85 10491455-3 1999 Exhaled carbon monoxide (CO), a product of heme degradation by heme oxygenase 1 (HO-1) which is induced by inflammatory cytokines and oxidants, was therefore tested as a non-invasive marker of airway inflammation and oxidative stress. Heme 43-47 heme oxygenase 1 Homo sapiens 81-85 10491455-10 1999 CONCLUSIONS: High exhaled CO concentrations in patients with cystic fibrosis may reflect induction of HO-1. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 102-106 10460761-5 1999 Tin protoporphyrin IX (50 micrometer) and zinc protoporphyrin IX (10 microM), inhibitors of HO-1, reduced hemin-induced and IL-1beta-induced inhibitory effects. tin protoporphyrin IX 0-21 heme oxygenase 1 Homo sapiens 92-96 10460761-5 1999 Tin protoporphyrin IX (50 micrometer) and zinc protoporphyrin IX (10 microM), inhibitors of HO-1, reduced hemin-induced and IL-1beta-induced inhibitory effects. zinc protoporphyrin 42-64 heme oxygenase 1 Homo sapiens 92-96 10460761-9 1999 These findings suggest that HO-1 induction provides protection against H(2)O(2)-induced injury of the cultured human airway epithelial cells in part via the HO-bilirubin pathway. Hydrogen Peroxide 71-79 heme oxygenase 1 Homo sapiens 28-32 10460761-9 1999 These findings suggest that HO-1 induction provides protection against H(2)O(2)-induced injury of the cultured human airway epithelial cells in part via the HO-bilirubin pathway. Bilirubin 160-169 heme oxygenase 1 Homo sapiens 28-32 12835114-3 1999 The effects of dopamine on HO-1 expression were inhibited by ascorbate implicating a free radical mechanism of action. Dopamine 15-23 heme oxygenase 1 Homo sapiens 27-31 12835114-3 1999 The effects of dopamine on HO-1 expression were inhibited by ascorbate implicating a free radical mechanism of action. Ascorbic Acid 61-70 heme oxygenase 1 Homo sapiens 27-31 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. Dopamine 0-8 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. Iron 31-35 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. tin mesoporphyrin 111-128 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. tin mesoporphyrin 130-134 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. Dexamethasone 139-152 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. Dexamethasone 154-157 heme oxygenase 1 Homo sapiens 175-179 12835114-4 1999 Dopamine-induced mitochondrial iron trapping was abrogated by administration of the heme oxygenase inhibitors, tin mesoporphyrin (SnMP) or dexamethasone (DEX) indicating that HO-1 upregulation is necessary for subsequent mitochondrial iron deposition in these cells. Iron 235-239 heme oxygenase 1 Homo sapiens 175-179 12835114-5 1999 Overexpression of the human HO-1 gene in cultured rat astroglia by transient transfection also stimulated mitochondrial (55)Fe deposition, an effect that was again preventible by SnMP or DEX administration. Iron 124-126 heme oxygenase 1 Homo sapiens 28-32 12835114-5 1999 Overexpression of the human HO-1 gene in cultured rat astroglia by transient transfection also stimulated mitochondrial (55)Fe deposition, an effect that was again preventible by SnMP or DEX administration. tin mesoporphyrin 179-183 heme oxygenase 1 Homo sapiens 28-32 12835114-5 1999 Overexpression of the human HO-1 gene in cultured rat astroglia by transient transfection also stimulated mitochondrial (55)Fe deposition, an effect that was again preventible by SnMP or DEX administration. Dexamethasone 187-190 heme oxygenase 1 Homo sapiens 28-32 12835114-6 1999 We hypothesize that free ferrous iron and carbon monoxide generated by HO-1-mediated heme degradation promote mitochondrial membrane injury and the deposition of redox-active iron within this organelle. ammonium ferrous sulfate 25-32 heme oxygenase 1 Homo sapiens 71-75 12835114-6 1999 We hypothesize that free ferrous iron and carbon monoxide generated by HO-1-mediated heme degradation promote mitochondrial membrane injury and the deposition of redox-active iron within this organelle. Iron 33-37 heme oxygenase 1 Homo sapiens 71-75 12835114-6 1999 We hypothesize that free ferrous iron and carbon monoxide generated by HO-1-mediated heme degradation promote mitochondrial membrane injury and the deposition of redox-active iron within this organelle. Carbon Monoxide 42-57 heme oxygenase 1 Homo sapiens 71-75 12835114-6 1999 We hypothesize that free ferrous iron and carbon monoxide generated by HO-1-mediated heme degradation promote mitochondrial membrane injury and the deposition of redox-active iron within this organelle. Heme 85-89 heme oxygenase 1 Homo sapiens 71-75 12835114-6 1999 We hypothesize that free ferrous iron and carbon monoxide generated by HO-1-mediated heme degradation promote mitochondrial membrane injury and the deposition of redox-active iron within this organelle. Iron 175-179 heme oxygenase 1 Homo sapiens 71-75 12835114-8 1999 Stress-induced up-regulation of HO-1 in astroglia may be responsible for the abnormal patterns of brain iron deposition and mitochondrial insufficiency documented in various human neurodegenerative disorders. Iron 104-108 heme oxygenase 1 Homo sapiens 32-36 10423162-0 1999 Induction of heme oxygenase-1 as a response in sensing the signals evoked by distinct nitric oxide donors. Nitric Oxide 86-98 heme oxygenase 1 Homo sapiens 13-29 10423162-1 1999 To gain insights into the cellular responses evoked by nitric oxide (NO), we have studied the effects of NO donors with distinct chemistries on the expression of heme oxygenase-1 mRNA by northern blot analysis. Nitric Oxide 55-67 heme oxygenase 1 Homo sapiens 162-178 10423162-2 1999 The expression levels of heme oxygenase-1 mRNA were increased significantly in DLD-1 human colorectal adenocarcinoma cells by treatment with each of three NO donors: sodium nitroprusside (SNP), S-nitroso-L-glutathione (GSNO), and 3-morpholinosydnonimine (SIN-1). Nitroprusside 166-186 heme oxygenase 1 Homo sapiens 25-41 10423162-2 1999 The expression levels of heme oxygenase-1 mRNA were increased significantly in DLD-1 human colorectal adenocarcinoma cells by treatment with each of three NO donors: sodium nitroprusside (SNP), S-nitroso-L-glutathione (GSNO), and 3-morpholinosydnonimine (SIN-1). S-Nitrosoglutathione 194-217 heme oxygenase 1 Homo sapiens 25-41 10423162-2 1999 The expression levels of heme oxygenase-1 mRNA were increased significantly in DLD-1 human colorectal adenocarcinoma cells by treatment with each of three NO donors: sodium nitroprusside (SNP), S-nitroso-L-glutathione (GSNO), and 3-morpholinosydnonimine (SIN-1). S-Nitrosoglutathione 219-223 heme oxygenase 1 Homo sapiens 25-41 10423162-2 1999 The expression levels of heme oxygenase-1 mRNA were increased significantly in DLD-1 human colorectal adenocarcinoma cells by treatment with each of three NO donors: sodium nitroprusside (SNP), S-nitroso-L-glutathione (GSNO), and 3-morpholinosydnonimine (SIN-1). linsidomine 230-253 heme oxygenase 1 Homo sapiens 25-41 10423162-3 1999 A combination of SIN-1 plus SNP or GSNO additively increased heme oxygenase-1 mRNA expression, whereas synergistic induction was seen with SNP plus GSNO. S-Nitrosoglutathione 35-39 heme oxygenase 1 Homo sapiens 61-77 10423162-6 1999 Transient transfection assays suggested that treatment with SNP, but not with GSNO or SIN-1, increased the expression of a reporter gene through a cis-acting element, including the cadmium-responsive element, of the human heme oxygenase-1 gene. Cadmium 181-188 heme oxygenase 1 Homo sapiens 222-238 10409239-2 1999 Overexpression of HO-1 in cells might, therefore, protect against oxidative stress produced by certain agents, specifically heme, by catalyzing its degradation to bilirubin, which by itself has antioxidant properties. Heme 124-128 heme oxygenase 1 Homo sapiens 18-22 10409239-2 1999 Overexpression of HO-1 in cells might, therefore, protect against oxidative stress produced by certain agents, specifically heme, by catalyzing its degradation to bilirubin, which by itself has antioxidant properties. Bilirubin 163-172 heme oxygenase 1 Homo sapiens 18-22 10409239-6 1999 Moreover, human HO-1 gene-transduced endothelial cells acquired substantial resistance to toxicity produced by exposure to heme and H(2)O(2) compared with that in nontransduced cells. Heme 123-127 heme oxygenase 1 Homo sapiens 16-20 10409239-7 1999 The protective effect of enhancement of HO-1 activity against heme and H(2)O(2) was reversed by pretreatment with stannic mesoporphyrin, a competitive inhibitor of HO. Heme 62-66 heme oxygenase 1 Homo sapiens 40-44 10409239-7 1999 The protective effect of enhancement of HO-1 activity against heme and H(2)O(2) was reversed by pretreatment with stannic mesoporphyrin, a competitive inhibitor of HO. Hydrogen Peroxide 71-79 heme oxygenase 1 Homo sapiens 40-44 10409239-7 1999 The protective effect of enhancement of HO-1 activity against heme and H(2)O(2) was reversed by pretreatment with stannic mesoporphyrin, a competitive inhibitor of HO. stannic mesoporphyrin 114-135 heme oxygenase 1 Homo sapiens 40-44 10403520-5 1999 The treatment of the two cell lines with 50 microM CdCl2 induced hsp72, hsp32 and metallothionein (MT-II) mRNAs, and the induction level of each mRNA did not differ in the two cell lines. Cadmium Chloride 51-56 heme oxygenase 1 Homo sapiens 72-77 10403520-6 1999 However, the treatment with 20 microM CdCl2 induced the hsp72 and hsp32 mRNAs in A2780CP cells less than in A2780 cells, while the MT-II mRNA was induced to similar levels in the two cell lines. Cadmium Chloride 38-43 heme oxygenase 1 Homo sapiens 66-71 10373419-1 1999 Whether or not reducing equivalents are indispensable for the conversion of ferric alpha-hydroxyheme bound to heme oxygenase-1 to verdoheme remains controversial (Matera, K. M., Takahashi, S., Fujii, H., Zhou, H., Ishikawa, K., Yoshimura, T., Rousseau, D. L., Yoshida, T., and Ikeda-Saito, M. (1996) J. Biol. ferric alpha-hydroxyheme 76-100 heme oxygenase 1 Homo sapiens 110-126 10513606-10 1999 Strain treatment, similar to H2O2 exposure, induced HO-1 expression in a time-dependent manner. Hydrogen Peroxide 29-33 heme oxygenase 1 Homo sapiens 52-56 10513606-12 1999 ECs treated with N-acetyl-cysteine abolished HO-1 gene induction. Acetylcysteine 17-34 heme oxygenase 1 Homo sapiens 45-49 10513606-13 1999 Our results suggest that cyclic strain-induced ROS cause a transient increase of glutathione peroxidase activity that results in a decrease of GSH level in ECs and that this decrease is crucial to HO-1 induction. Reactive Oxygen Species 47-50 heme oxygenase 1 Homo sapiens 197-201 10373419-1 1999 Whether or not reducing equivalents are indispensable for the conversion of ferric alpha-hydroxyheme bound to heme oxygenase-1 to verdoheme remains controversial (Matera, K. M., Takahashi, S., Fujii, H., Zhou, H., Ishikawa, K., Yoshimura, T., Rousseau, D. L., Yoshida, T., and Ikeda-Saito, M. (1996) J. Biol. verdoheme 130-139 heme oxygenase 1 Homo sapiens 110-126 10373419-6 1999 To resolve this controversy, we have prepared a ferric alpha-hydroxyheme-heme oxygenase-1 complex and titrated the complex with O2 under strictly anaerobic conditions. Oxygen 128-130 heme oxygenase 1 Homo sapiens 73-89 10373419-11 1999 The verdoheme formed from alpha-hydroxyheme was shown to be in the ferrous oxidation state by the addition of CO or potassium ferricyanide to the resultant verdoheme-heme oxygenase-1 complex. verdoheme 4-13 heme oxygenase 1 Homo sapiens 166-182 10373419-11 1999 The verdoheme formed from alpha-hydroxyheme was shown to be in the ferrous oxidation state by the addition of CO or potassium ferricyanide to the resultant verdoheme-heme oxygenase-1 complex. alpha-hydroxyheme 26-43 heme oxygenase 1 Homo sapiens 166-182 10373419-11 1999 The verdoheme formed from alpha-hydroxyheme was shown to be in the ferrous oxidation state by the addition of CO or potassium ferricyanide to the resultant verdoheme-heme oxygenase-1 complex. Carbon Monoxide 110-112 heme oxygenase 1 Homo sapiens 166-182 10373419-11 1999 The verdoheme formed from alpha-hydroxyheme was shown to be in the ferrous oxidation state by the addition of CO or potassium ferricyanide to the resultant verdoheme-heme oxygenase-1 complex. potassium ferricyanide 116-138 heme oxygenase 1 Homo sapiens 166-182 10330231-4 1999 Acute exposure to the PKC activator phorbol 12-myristate 13-acetate (PMA) stimulated HO-1 mRNA. Tetradecanoylphorbol Acetate 36-67 heme oxygenase 1 Homo sapiens 85-89 10349844-8 1999 Moreover, pretreatment with interferon-gamma partially suppressed the induction of heme oxygenase-1 mRNA expression caused by either sodium nitroprusside, cadmium, or hemin. Nitroprusside 133-153 heme oxygenase 1 Homo sapiens 83-99 10349844-8 1999 Moreover, pretreatment with interferon-gamma partially suppressed the induction of heme oxygenase-1 mRNA expression caused by either sodium nitroprusside, cadmium, or hemin. Cadmium 155-162 heme oxygenase 1 Homo sapiens 83-99 10349844-8 1999 Moreover, pretreatment with interferon-gamma partially suppressed the induction of heme oxygenase-1 mRNA expression caused by either sodium nitroprusside, cadmium, or hemin. Hemin 167-172 heme oxygenase 1 Homo sapiens 83-99 10330231-4 1999 Acute exposure to the PKC activator phorbol 12-myristate 13-acetate (PMA) stimulated HO-1 mRNA. Tetradecanoylphorbol Acetate 69-72 heme oxygenase 1 Homo sapiens 85-89 10330231-6 1999 Additionally, the phosphatase inhibitors okadaic acid and calyculin enhanced cytokine induction of HO-1. Okadaic Acid 41-53 heme oxygenase 1 Homo sapiens 99-103 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Quinacrine 0-9 heme oxygenase 1 Homo sapiens 39-43 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Quinacrine 0-9 heme oxygenase 1 Homo sapiens 155-159 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Arachidonic Acid 89-101 heme oxygenase 1 Homo sapiens 39-43 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Arachidonic Acid 89-101 heme oxygenase 1 Homo sapiens 155-159 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Phospholipids 137-150 heme oxygenase 1 Homo sapiens 39-43 10330231-7 1999 Mepacrine, a PLA2 inhibitor, prevented HO-1 induction by cytokine, suggesting a role for arachidonate, the product of PLA2 hydrolysis of phospholipids, in HO-1 expression. Phospholipids 137-150 heme oxygenase 1 Homo sapiens 155-159 10330231-8 1999 The intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N",N"-tetraacetic acid acetoxymethyl ester (BAPTA-AM) blocked cytokine induction of HO-1. Calcium 18-25 heme oxygenase 1 Homo sapiens 153-157 10330231-8 1999 The intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N",N"-tetraacetic acid acetoxymethyl ester (BAPTA-AM) blocked cytokine induction of HO-1. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 35-111 heme oxygenase 1 Homo sapiens 153-157 10330231-8 1999 The intracellular calcium chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N",N"-tetraacetic acid acetoxymethyl ester (BAPTA-AM) blocked cytokine induction of HO-1. 1,2-bis(2-aminophenoxy)ethane N,N,N',N'-tetraacetic acid acetoxymethyl ester 113-121 heme oxygenase 1 Homo sapiens 153-157 10330231-9 1999 Paradoxically, the calcium ionophore A-23187 prevented HO-1 induction by cytokine but not by PMA. Calcium 19-26 heme oxygenase 1 Homo sapiens 55-59 10330231-10 1999 Finally, the oxidant scavenger N-acetylcysteine inhibited HO-1 induction by cytokines. Acetylcysteine 31-47 heme oxygenase 1 Homo sapiens 58-62 10217256-3 1999 We also provide evidence that up-regulation of the stress protein heme oxygenase-1 (HO-1) is both necessary and sufficient for mitochondrial iron trapping in dopamine-challenged astroglia. Dopamine 158-166 heme oxygenase 1 Homo sapiens 66-82 10425949-2 1999 Synthesis of the enzyme heme oxygenase-1 (HO-1) is increased after exposure of eukaryotic cells to conditions of oxidative stress (including H2O2) as a defense against the damaging effects of free radicals. Hydrogen Peroxide 141-145 heme oxygenase 1 Homo sapiens 24-40 10217256-3 1999 We also provide evidence that up-regulation of the stress protein heme oxygenase-1 (HO-1) is both necessary and sufficient for mitochondrial iron trapping in dopamine-challenged astroglia. Iron 141-145 heme oxygenase 1 Homo sapiens 66-82 10101013-0 1999 Iron regulates hyperoxia-dependent human heme oxygenase 1 gene expression in pulmonary endothelial cells. Iron 0-4 heme oxygenase 1 Homo sapiens 41-57 10101013-3 1999 Recently, heme oxygenase 1 (HO-1), the rate-limiting enzyme in the metabolism of heme, has been found to have a protective role in oxidant injury. Heme 10-14 heme oxygenase 1 Homo sapiens 28-32 10101013-8 1999 We also found that HO-1 expression depended on chelatable iron. Iron 58-62 heme oxygenase 1 Homo sapiens 19-23 10101013-9 1999 The iron chelator desferrioxamine not only inhibited the iron- dependent potentiation of HO-1 in response to hyperoxia but also inhibited both hyperoxia and basal expression. Iron 4-8 heme oxygenase 1 Homo sapiens 89-93 10101013-9 1999 The iron chelator desferrioxamine not only inhibited the iron- dependent potentiation of HO-1 in response to hyperoxia but also inhibited both hyperoxia and basal expression. Deferoxamine 18-33 heme oxygenase 1 Homo sapiens 89-93 10101013-9 1999 The iron chelator desferrioxamine not only inhibited the iron- dependent potentiation of HO-1 in response to hyperoxia but also inhibited both hyperoxia and basal expression. Iron 57-61 heme oxygenase 1 Homo sapiens 89-93 10101013-11 1999 We also evaluated 4.5 kb of the human HO-1 promoter region and demonstrated that this region has promoter activity to the stimulus heme; however, there was no evidence of promoter activity to either iron or hyperoxia. Heme 131-135 heme oxygenase 1 Homo sapiens 38-42 10101013-12 1999 This diversity of promoter activity to heme, heavy metals, and hyperoxia is unique to the human HO-1 gene. Heme 39-43 heme oxygenase 1 Homo sapiens 96-100 10425949-2 1999 Synthesis of the enzyme heme oxygenase-1 (HO-1) is increased after exposure of eukaryotic cells to conditions of oxidative stress (including H2O2) as a defense against the damaging effects of free radicals. Hydrogen Peroxide 141-145 heme oxygenase 1 Homo sapiens 42-46 10425949-3 1999 Dental pulps were evaluated for HO-1 (aka Heat Shock Protein 32) presence in teeth treated with 10% carbamide peroxide. Carbamide Peroxide 100-118 heme oxygenase 1 Homo sapiens 32-36 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Iron 83-87 heme oxygenase 1 Homo sapiens 35-51 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Iron 83-87 heme oxygenase 1 Homo sapiens 53-58 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Cysteine 112-120 heme oxygenase 1 Homo sapiens 35-51 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Cysteine 112-120 heme oxygenase 1 Homo sapiens 53-58 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Tyrosine 124-132 heme oxygenase 1 Homo sapiens 35-51 10090762-1 1999 The H25C and H25Y mutants of human heme oxygenase-1 (hHO-1), in which the proximal iron ligand is replaced by a cysteine or tyrosine, have been expressed and characterized. Tyrosine 124-132 heme oxygenase 1 Homo sapiens 53-58 10218639-4 1999 We also demonstrate a high degree of correlation between the amount of heme released and the degree of subsequent induction of heme oxygenase 1 transcription following UVA and hydrogen peroxide treatment. Heme 71-75 heme oxygenase 1 Homo sapiens 127-143 10218639-0 1999 Cyclooxygenase dependent release of heme from microsomal hemeproteins correlates with induction of heme oxygenase 1 transcription in human fibroblasts. Heme 36-40 heme oxygenase 1 Homo sapiens 99-115 10218639-4 1999 We also demonstrate a high degree of correlation between the amount of heme released and the degree of subsequent induction of heme oxygenase 1 transcription following UVA and hydrogen peroxide treatment. Hydrogen Peroxide 176-193 heme oxygenase 1 Homo sapiens 127-143 10218639-1 1999 Induction of heme oxygenase 1 transcription and enzymatic activity is a common response after exposure of cells to various forms of oxidative stress including ultraviolet A radiation (UVA) and hydrogen peroxide. Hydrogen Peroxide 193-210 heme oxygenase 1 Homo sapiens 13-29 10218639-5 1999 We propose that release of heme from microsomal hemeproteins determines the degree of induction of heme oxygenase 1 transcription in human fibroblasts after oxidative stress. Heme 27-31 heme oxygenase 1 Homo sapiens 99-115 9920842-5 1999 Because HO-1 lysed peptide-loaded Fas-deficient target cells derived from a patient with a homozygous Fas gene mutation, its cytotoxicity appeared to be mediated by a Fas-independent pathway. ammonium ferrous sulfate 34-37 heme oxygenase 1 Homo sapiens 8-12 9920842-5 1999 Because HO-1 lysed peptide-loaded Fas-deficient target cells derived from a patient with a homozygous Fas gene mutation, its cytotoxicity appeared to be mediated by a Fas-independent pathway. ammonium ferrous sulfate 102-105 heme oxygenase 1 Homo sapiens 8-12 9920842-8 1999 HO-1 expressed mRNA for apoptosis-inducing mediators, including perforin, granzyme B, Fas ligand, TNF-alpha, and lymphotoxin; however, no DNA fragmentation was detected in target cells incubated with HO-1 by 5-[125I]Iodo-2"-deoxyuridine release assay and agarose gel electrophoresis of DNA. 5-[125i]iodo-2"-deoxyuridine 208-236 heme oxygenase 1 Homo sapiens 0-4 9920842-8 1999 HO-1 expressed mRNA for apoptosis-inducing mediators, including perforin, granzyme B, Fas ligand, TNF-alpha, and lymphotoxin; however, no DNA fragmentation was detected in target cells incubated with HO-1 by 5-[125I]Iodo-2"-deoxyuridine release assay and agarose gel electrophoresis of DNA. Sepharose 255-262 heme oxygenase 1 Homo sapiens 0-4 9920842-9 1999 Although it has been suggested that the Fas/Fas ligand system is the main pathway by which CD4(+) CTL-mediated cytotoxicity is exerted in murine systems, HO-1 produced peptide-specific and HLA-restricted cytotoxicity via a Fas-independent and nonapoptotic pathway. ammonium ferrous sulfate 40-43 heme oxygenase 1 Homo sapiens 154-158 9920842-9 1999 Although it has been suggested that the Fas/Fas ligand system is the main pathway by which CD4(+) CTL-mediated cytotoxicity is exerted in murine systems, HO-1 produced peptide-specific and HLA-restricted cytotoxicity via a Fas-independent and nonapoptotic pathway. ammonium ferrous sulfate 44-47 heme oxygenase 1 Homo sapiens 154-158 11089885-11 1999 The expression of heme oxygenase (HO-1), a stress-response protein, has been used to monitor effects of hyperthermia, 12-O-tetradecanoly phorbol 13-acetate (TPA) and 4-HNE. 12-o-tetradecanoly phorbol 13-acetate 118-155 heme oxygenase 1 Homo sapiens 34-38 10083452-0 1999 Glutamine-induced heme oxygenase-1 protects intestines and hearts from warm ischemic injury. Glutamine 0-9 heme oxygenase 1 Homo sapiens 18-34 10943654-1 1999 Oxidative stress and hypoxia, which may occur in cystic fibrosis patients (CF) at rest and may be worsened by exercise, induce the expression of heme oxygenase (HO)-1, resulting in increased carbon monoxide (CO) formation. Carbon Monoxide 191-206 heme oxygenase 1 Homo sapiens 145-166 10943654-1 1999 Oxidative stress and hypoxia, which may occur in cystic fibrosis patients (CF) at rest and may be worsened by exercise, induce the expression of heme oxygenase (HO)-1, resulting in increased carbon monoxide (CO) formation. Carbon Monoxide 208-210 heme oxygenase 1 Homo sapiens 145-166 11089885-11 1999 The expression of heme oxygenase (HO-1), a stress-response protein, has been used to monitor effects of hyperthermia, 12-O-tetradecanoly phorbol 13-acetate (TPA) and 4-HNE. Tetradecanoylphorbol Acetate 157-160 heme oxygenase 1 Homo sapiens 34-38 11089885-11 1999 The expression of heme oxygenase (HO-1), a stress-response protein, has been used to monitor effects of hyperthermia, 12-O-tetradecanoly phorbol 13-acetate (TPA) and 4-HNE. 4-hydroxy-2-nonenal 166-171 heme oxygenase 1 Homo sapiens 34-38 9808084-0 1998 Linoleyl hydroperoxide transcriptionally upregulates heme oxygenase-1 gene expression in human renal epithelial and aortic endothelial cells. linoleyl hydroperoxide 0-22 heme oxygenase 1 Homo sapiens 53-69 9890653-1 1999 Inducible heme oxygenase (HO-1) is an antioxidant stress protein, that is mainly induced by reactive oxygen species (ROS), cytokines and hyperthermia. Reactive Oxygen Species 92-115 heme oxygenase 1 Homo sapiens 26-30 9890653-1 1999 Inducible heme oxygenase (HO-1) is an antioxidant stress protein, that is mainly induced by reactive oxygen species (ROS), cytokines and hyperthermia. Reactive Oxygen Species 117-120 heme oxygenase 1 Homo sapiens 26-30 9884071-1 1998 The contribution of haeme oxygenase-derived carbon monoxide (CO) to the regulation of vascular tone in thoracic aorta was investigated following induction of the inducible isoform of haeme oxygenase (HO-1). Carbon Monoxide 61-63 heme oxygenase 1 Homo sapiens 200-204 9884071-6 1998 Induction of HO-1 in SNAP-treated rings was associated with a higher 14CO release compared to control, as measured by scintillation counting after incubation of aortas with [2-14C]-L-glycine, the precursor of haeme. 14co 69-73 heme oxygenase 1 Homo sapiens 13-17 9884071-6 1998 Induction of HO-1 in SNAP-treated rings was associated with a higher 14CO release compared to control, as measured by scintillation counting after incubation of aortas with [2-14C]-L-glycine, the precursor of haeme. [2-14c]-l-glycine 173-190 heme oxygenase 1 Homo sapiens 13-17 9884071-7 1998 Guanosine 3",5"-monophosphate (cyclic GMP) content was also greatly enhanced in aortas expressing high levels of HO-1. Cyclic GMP 0-29 heme oxygenase 1 Homo sapiens 113-117 9863766-8 1998 Concurrent with these changes, mercuric chloride treatment activates transcription of the heme oxygenase-1 (HO-1) gene in a dose dependent manner, further indicating an oxidative stress response. Mercuric Chloride 31-48 heme oxygenase 1 Homo sapiens 90-106 9863766-8 1998 Concurrent with these changes, mercuric chloride treatment activates transcription of the heme oxygenase-1 (HO-1) gene in a dose dependent manner, further indicating an oxidative stress response. Mercuric Chloride 31-48 heme oxygenase 1 Homo sapiens 108-112 9808084-3 1998 Oxidized LDL has been shown to induce heme oxygenase-1 (HO-1), a microsomal enzyme that is involved in heme detoxification and is a major endogenous source of carbon monoxide. Carbon Monoxide 159-174 heme oxygenase 1 Homo sapiens 38-54 9808084-3 1998 Oxidized LDL has been shown to induce heme oxygenase-1 (HO-1), a microsomal enzyme that is involved in heme detoxification and is a major endogenous source of carbon monoxide. Carbon Monoxide 159-174 heme oxygenase 1 Homo sapiens 56-60 9808084-6 1998 Exposure to LAox (25 microM) showed an approximately 16-fold induction of HO-1 mRNA, whereas exposure to lyso-PC (25 microM) showed only an approximate 2.6-fold increase. laox 12-16 heme oxygenase 1 Homo sapiens 74-78 9808084-7 1998 Treatment with actinomycin-D (4 microM), a transcriptional inhibitor, as well as nuclear run-on assays, demonstrated that LAox-mediated HO-1 gene induction is dependent on de novo transcription. Dactinomycin 15-28 heme oxygenase 1 Homo sapiens 136-140 9808084-7 1998 Treatment with actinomycin-D (4 microM), a transcriptional inhibitor, as well as nuclear run-on assays, demonstrated that LAox-mediated HO-1 gene induction is dependent on de novo transcription. laox 122-126 heme oxygenase 1 Homo sapiens 136-140 9808084-10 1998 These studies are the first demonstration that LAox induces HO-1 by transcriptional mechanisms and may have implications in the pathogenesis of cell injury in atherosclerosis and progressive renal disease. laox 47-51 heme oxygenase 1 Homo sapiens 60-64 10193374-2 1998 Induction of heme oxygenase (HO-1) by reactive oxygen species is a general cytoprotective mechanism against oxidative stress. Reactive Oxygen Species 38-61 heme oxygenase 1 Homo sapiens 29-33 10193374-3 1998 HO-1 catabolises heme to bilirubin, free iron and carbon monoxide (CO). Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 10193374-3 1998 HO-1 catabolises heme to bilirubin, free iron and carbon monoxide (CO). Bilirubin 25-34 heme oxygenase 1 Homo sapiens 0-4 10193374-3 1998 HO-1 catabolises heme to bilirubin, free iron and carbon monoxide (CO). Iron 41-45 heme oxygenase 1 Homo sapiens 0-4 10193374-3 1998 HO-1 catabolises heme to bilirubin, free iron and carbon monoxide (CO). Carbon Monoxide 50-65 heme oxygenase 1 Homo sapiens 0-4 10193374-3 1998 HO-1 catabolises heme to bilirubin, free iron and carbon monoxide (CO). Carbon Monoxide 67-69 heme oxygenase 1 Homo sapiens 0-4 9784401-0 1998 Human lymphocyte heme oxygenase 1 as a response biomarker to inorganic arsenic. Arsenic 71-78 heme oxygenase 1 Homo sapiens 17-33 9784401-2 1998 We report the induction of HO1 in human lymphoblastoid cells (LBs) by arsenite in a dose-related manner. arsenite 70-78 heme oxygenase 1 Homo sapiens 27-30 9784401-1 1998 We propose the use of human lymphocyte heme oxygenase 1 (HO1) as a biomarker of response to environmental arsenic exposure. Arsenic 106-113 heme oxygenase 1 Homo sapiens 39-55 9784401-3 1998 HO1 was identified by SDS-PAGE from its molecular weight and from its detection by Western blotting with anti-HO1. Sodium Dodecyl Sulfate 22-25 heme oxygenase 1 Homo sapiens 0-3 9784401-1 1998 We propose the use of human lymphocyte heme oxygenase 1 (HO1) as a biomarker of response to environmental arsenic exposure. Arsenic 106-113 heme oxygenase 1 Homo sapiens 57-60 9784401-4 1998 HO1 levels in LBs treated with arsenite increased by de novo synthesis as demonstrated by incorporation of 35S-methionine and by inhibition of HO1 synthesis by actinomycin D. arsenite 31-39 heme oxygenase 1 Homo sapiens 0-3 9784401-4 1998 HO1 levels in LBs treated with arsenite increased by de novo synthesis as demonstrated by incorporation of 35S-methionine and by inhibition of HO1 synthesis by actinomycin D. arsenite 31-39 heme oxygenase 1 Homo sapiens 143-146 9722676-0 1998 Involvement of the tyrosine phosphorylation pathway in induction of human heme oxygenase-1 by hemin, sodium arsenite, and cadmium chloride. Tyrosine 19-27 heme oxygenase 1 Homo sapiens 74-90 9784401-4 1998 HO1 levels in LBs treated with arsenite increased by de novo synthesis as demonstrated by incorporation of 35S-methionine and by inhibition of HO1 synthesis by actinomycin D. 35s-methionine 107-121 heme oxygenase 1 Homo sapiens 0-3 9784401-4 1998 HO1 levels in LBs treated with arsenite increased by de novo synthesis as demonstrated by incorporation of 35S-methionine and by inhibition of HO1 synthesis by actinomycin D. Dactinomycin 160-173 heme oxygenase 1 Homo sapiens 0-3 9784401-4 1998 HO1 levels in LBs treated with arsenite increased by de novo synthesis as demonstrated by incorporation of 35S-methionine and by inhibition of HO1 synthesis by actinomycin D. Dactinomycin 160-173 heme oxygenase 1 Homo sapiens 143-146 9784401-6 1998 HO1 was also induced by 10 microM cadmium or mercuric chloride. Cadmium 34-41 heme oxygenase 1 Homo sapiens 0-3 9784401-6 1998 HO1 was also induced by 10 microM cadmium or mercuric chloride. Mercuric Chloride 45-62 heme oxygenase 1 Homo sapiens 0-3 9784401-7 1998 We suggest that circulating lymphocyte HO1 levels may be useful in assessing the biological activity of arsenic exposure in vivo under properly controlled conditions of simultaneous urinalysis for arsenic, cadmium, and mercury. Arsenic 104-111 heme oxygenase 1 Homo sapiens 39-42 9784401-7 1998 We suggest that circulating lymphocyte HO1 levels may be useful in assessing the biological activity of arsenic exposure in vivo under properly controlled conditions of simultaneous urinalysis for arsenic, cadmium, and mercury. Arsenic 197-204 heme oxygenase 1 Homo sapiens 39-42 9784401-7 1998 We suggest that circulating lymphocyte HO1 levels may be useful in assessing the biological activity of arsenic exposure in vivo under properly controlled conditions of simultaneous urinalysis for arsenic, cadmium, and mercury. Cadmium 206-213 heme oxygenase 1 Homo sapiens 39-42 9784401-7 1998 We suggest that circulating lymphocyte HO1 levels may be useful in assessing the biological activity of arsenic exposure in vivo under properly controlled conditions of simultaneous urinalysis for arsenic, cadmium, and mercury. Mercury 219-226 heme oxygenase 1 Homo sapiens 39-42 9722676-4 1998 Genistein (50 microM), another tyrosine kinase inhibitor, also inhibited the induction of HO-1 mRNA by hemin, arsenite, and cadmium. Hemin 103-108 heme oxygenase 1 Homo sapiens 90-94 9722676-4 1998 Genistein (50 microM), another tyrosine kinase inhibitor, also inhibited the induction of HO-1 mRNA by hemin, arsenite, and cadmium. arsenite 110-118 heme oxygenase 1 Homo sapiens 90-94 9722676-4 1998 Genistein (50 microM), another tyrosine kinase inhibitor, also inhibited the induction of HO-1 mRNA by hemin, arsenite, and cadmium. Cadmium 124-131 heme oxygenase 1 Homo sapiens 90-94 9747510-0 1998 Oxygen tension regulates heme oxygenase-1 gene expression in mammalian cell lines. Oxygen 0-6 heme oxygenase 1 Homo sapiens 25-41 9747510-5 1998 Cycloheximide and actinomycin D inhibited the increases in the HO-1 mRNA level produced by hyperoxia, indicating that response to hyperoxia is dependent on de novo protein synthesis and mRNA transcription. Cycloheximide 0-13 heme oxygenase 1 Homo sapiens 63-67 9747510-5 1998 Cycloheximide and actinomycin D inhibited the increases in the HO-1 mRNA level produced by hyperoxia, indicating that response to hyperoxia is dependent on de novo protein synthesis and mRNA transcription. Dactinomycin 18-31 heme oxygenase 1 Homo sapiens 63-67 9747510-6 1998 Antioxidants, desferrioxamine (DES) and o-phenanthroline (OP) partially inhibited the HO-1 mRNA elevation by hyperoxia. Deferoxamine 14-29 heme oxygenase 1 Homo sapiens 86-90 9747510-6 1998 Antioxidants, desferrioxamine (DES) and o-phenanthroline (OP) partially inhibited the HO-1 mRNA elevation by hyperoxia. Deferoxamine 31-34 heme oxygenase 1 Homo sapiens 86-90 9747510-6 1998 Antioxidants, desferrioxamine (DES) and o-phenanthroline (OP) partially inhibited the HO-1 mRNA elevation by hyperoxia. 1,10-phenanthroline 40-56 heme oxygenase 1 Homo sapiens 86-90 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. sodium arsenite 26-41 heme oxygenase 1 Homo sapiens 181-185 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. sodium arsenite 43-49 heme oxygenase 1 Homo sapiens 181-185 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. Cadmium Chloride 52-68 heme oxygenase 1 Homo sapiens 181-185 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. cdcl 70-74 heme oxygenase 1 Homo sapiens 181-185 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. Hydrogen Peroxide 83-100 heme oxygenase 1 Homo sapiens 181-185 9747510-7 1998 In addition to hyperoxia, sodium arsenite (NaAsO2), cadmium chloride (CdCl(2)) and hydrogen peroxide (H2O2), which are reactive oxygen intermediates (ROI) generators, increased the HO-1 mRNA level by 11-, 22- and 2.5-fold, respectively. Hydrogen Peroxide 102-106 heme oxygenase 1 Homo sapiens 181-185 9747510-9 1998 In contrast to OP, N-acetylcysteine (NAC), an antioxidant and membrane-permeable reducing reagent, enhanced the HO-1 mRNA elevation induced by hyperoxia, although NAC inhibited the mRNA elevation induced by NaAsO2, CdCl2 and H2O2. Acetylcysteine 19-35 heme oxygenase 1 Homo sapiens 112-116 9747510-9 1998 In contrast to OP, N-acetylcysteine (NAC), an antioxidant and membrane-permeable reducing reagent, enhanced the HO-1 mRNA elevation induced by hyperoxia, although NAC inhibited the mRNA elevation induced by NaAsO2, CdCl2 and H2O2. Acetylcysteine 37-40 heme oxygenase 1 Homo sapiens 112-116 9747510-10 1998 These results indicate that oxygen tension regulates HO-1 gene expression and suggest that hyperoxia-specific and redox-sensitive regulators may be involved in hyperoxia-mediated HO-1 gene expression. Oxygen 28-34 heme oxygenase 1 Homo sapiens 53-57 9747510-10 1998 These results indicate that oxygen tension regulates HO-1 gene expression and suggest that hyperoxia-specific and redox-sensitive regulators may be involved in hyperoxia-mediated HO-1 gene expression. Oxygen 28-34 heme oxygenase 1 Homo sapiens 179-183 9855696-5 1998 In contrast, the HO-1 isoform is strongly induced in vascular cells by various stress-associated agents and markedly increases CO synthesis during pathological conditions. Carbon Monoxide 127-129 heme oxygenase 1 Homo sapiens 17-21 9722676-5 1998 Nuclear runoff assays revealed that herbimycin blocked the hemin-induced transcription of the HO-1 gene. herbimycin 36-46 heme oxygenase 1 Homo sapiens 94-98 9722676-5 1998 Nuclear runoff assays revealed that herbimycin blocked the hemin-induced transcription of the HO-1 gene. Hemin 59-64 heme oxygenase 1 Homo sapiens 94-98 9722676-6 1998 The induction of HO-1 mRNA by hemin in human peripheral blood mononuclear cells was inhibited by herbimycin. herbimycin 97-107 heme oxygenase 1 Homo sapiens 17-21 9722676-8 1998 When HeLa cells were treated with a specific inhibitor of the mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase cascade, PD58059 (100 microM), suppression of the cadmium-dependent HO-1 induction was not observed, but the hemin- or arsenite-dependent induction was slightly inhibited. pd58059 149-156 heme oxygenase 1 Homo sapiens 208-212 9722676-8 1998 When HeLa cells were treated with a specific inhibitor of the mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase cascade, PD58059 (100 microM), suppression of the cadmium-dependent HO-1 induction was not observed, but the hemin- or arsenite-dependent induction was slightly inhibited. Cadmium 190-197 heme oxygenase 1 Homo sapiens 208-212 9722676-8 1998 When HeLa cells were treated with a specific inhibitor of the mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase cascade, PD58059 (100 microM), suppression of the cadmium-dependent HO-1 induction was not observed, but the hemin- or arsenite-dependent induction was slightly inhibited. Hemin 249-254 heme oxygenase 1 Homo sapiens 208-212 9722676-8 1998 When HeLa cells were treated with a specific inhibitor of the mitogen-activated protein kinase (MAPK)/extracellular-signal regulated kinase cascade, PD58059 (100 microM), suppression of the cadmium-dependent HO-1 induction was not observed, but the hemin- or arsenite-dependent induction was slightly inhibited. arsenite 259-267 heme oxygenase 1 Homo sapiens 208-212 9722676-0 1998 Involvement of the tyrosine phosphorylation pathway in induction of human heme oxygenase-1 by hemin, sodium arsenite, and cadmium chloride. Hemin 94-99 heme oxygenase 1 Homo sapiens 74-90 9722676-0 1998 Involvement of the tyrosine phosphorylation pathway in induction of human heme oxygenase-1 by hemin, sodium arsenite, and cadmium chloride. sodium arsenite 101-116 heme oxygenase 1 Homo sapiens 74-90 9722676-0 1998 Involvement of the tyrosine phosphorylation pathway in induction of human heme oxygenase-1 by hemin, sodium arsenite, and cadmium chloride. Cadmium Chloride 122-138 heme oxygenase 1 Homo sapiens 74-90 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. herbimycin 43-55 heme oxygenase 1 Homo sapiens 77-93 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. herbimycin 43-55 heme oxygenase 1 Homo sapiens 95-99 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. Hemin 137-142 heme oxygenase 1 Homo sapiens 77-93 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. Hemin 137-142 heme oxygenase 1 Homo sapiens 95-99 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. sodium arsenite 144-159 heme oxygenase 1 Homo sapiens 77-93 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. sodium arsenite 144-159 heme oxygenase 1 Homo sapiens 95-99 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. Cadmium Chloride 164-180 heme oxygenase 1 Homo sapiens 77-93 9722676-1 1998 The effect of a tyrosine kinase inhibitor, herbimycin A, on the induction of heme oxygenase-1 (HO-1) mRNA in HeLa cells upon exposure to hemin, sodium arsenite and cadmium chloride was examined. Cadmium Chloride 164-180 heme oxygenase 1 Homo sapiens 95-99 9722676-2 1998 The induction of HO-1 mRNA by hemin was inhibited when the cells were pretreated with herbimycin A. Hemin 30-35 heme oxygenase 1 Homo sapiens 17-21 9722676-2 1998 The induction of HO-1 mRNA by hemin was inhibited when the cells were pretreated with herbimycin A. herbimycin 86-98 heme oxygenase 1 Homo sapiens 17-21 9722676-3 1998 Herbimycin also inhibited arsenite- and cadmium-dependent induction of HO-1 mRNA in a dose-dependent manner, but less inhibition was observed in cadmium-treated cells than in ones treated with hemin- or arsenite. herbimycin 0-10 heme oxygenase 1 Homo sapiens 71-75 9722676-3 1998 Herbimycin also inhibited arsenite- and cadmium-dependent induction of HO-1 mRNA in a dose-dependent manner, but less inhibition was observed in cadmium-treated cells than in ones treated with hemin- or arsenite. arsenite 26-34 heme oxygenase 1 Homo sapiens 71-75 9722676-3 1998 Herbimycin also inhibited arsenite- and cadmium-dependent induction of HO-1 mRNA in a dose-dependent manner, but less inhibition was observed in cadmium-treated cells than in ones treated with hemin- or arsenite. Cadmium 40-47 heme oxygenase 1 Homo sapiens 71-75 9722676-4 1998 Genistein (50 microM), another tyrosine kinase inhibitor, also inhibited the induction of HO-1 mRNA by hemin, arsenite, and cadmium. Genistein 0-9 heme oxygenase 1 Homo sapiens 90-94 9828853-0 1998 Raised levels of exhaled carbon monoxide are associated with an increased expression of heme oxygenase-1 in airway macrophages in asthma: a new marker of oxidative stress. Carbon Monoxide 25-40 heme oxygenase 1 Homo sapiens 88-104 9766835-4 1998 NAC treatment raised nerve glutathione levels compared to untreated nerves, as indicated using hemeoxygenase-1 (hsp32) immunoreactivity as a marker of glutathione depletion. Acetylcysteine 0-3 heme oxygenase 1 Homo sapiens 112-117 9828853-3 1998 HO-1 catabolises heme to bilirubin, free iron, and carbon monoxide (CO). Heme 17-21 heme oxygenase 1 Homo sapiens 0-4 9828853-3 1998 HO-1 catabolises heme to bilirubin, free iron, and carbon monoxide (CO). Bilirubin 25-34 heme oxygenase 1 Homo sapiens 0-4 9828853-3 1998 HO-1 catabolises heme to bilirubin, free iron, and carbon monoxide (CO). Iron 41-45 heme oxygenase 1 Homo sapiens 0-4 9828853-3 1998 HO-1 catabolises heme to bilirubin, free iron, and carbon monoxide (CO). Carbon Monoxide 51-66 heme oxygenase 1 Homo sapiens 0-4 9828853-3 1998 HO-1 catabolises heme to bilirubin, free iron, and carbon monoxide (CO). Carbon Monoxide 68-70 heme oxygenase 1 Homo sapiens 0-4 9828853-9 1998 CONCLUSIONS: Increased exhaled CO levels and HO-1 expression may reflect induction of HO-1 which may be inhibited by steroids. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 45-49 9828853-9 1998 CONCLUSIONS: Increased exhaled CO levels and HO-1 expression may reflect induction of HO-1 which may be inhibited by steroids. Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 86-90 9828853-9 1998 CONCLUSIONS: Increased exhaled CO levels and HO-1 expression may reflect induction of HO-1 which may be inhibited by steroids. Steroids 117-125 heme oxygenase 1 Homo sapiens 45-49 9828853-9 1998 CONCLUSIONS: Increased exhaled CO levels and HO-1 expression may reflect induction of HO-1 which may be inhibited by steroids. Steroids 117-125 heme oxygenase 1 Homo sapiens 86-90 9486963-0 1998 Hemeoxygenase-1 inhibits human myometrial contractility via carbon monoxide and is upregulated by progesterone during pregnancy. Carbon Monoxide 60-75 heme oxygenase 1 Homo sapiens 0-15 9622162-5 1998 This upregulation of HO-1 was completely blocked by the antioxidant N-acetylcysteine (NAC, 20 mmol/L). Acetylcysteine 68-84 heme oxygenase 1 Homo sapiens 21-25 9622162-5 1998 This upregulation of HO-1 was completely blocked by the antioxidant N-acetylcysteine (NAC, 20 mmol/L). Acetylcysteine 86-89 heme oxygenase 1 Homo sapiens 21-25 9622162-6 1998 In contrast, steady laminar shear (5 dyne/cm2) induced NADH oxidase activity and NAC-sensitive HO-1 mRNA expression only at 1 and 5 hours, a transient response that returned toward baseline at 24 hours. Acetylcysteine 81-84 heme oxygenase 1 Homo sapiens 95-99 9545559-9 1998 Furthermore, this low concentration of NAC also decreased the CdCl2-induced gene expression of HSP32 which represents a general response to oxidative stress. Cadmium Chloride 62-67 heme oxygenase 1 Homo sapiens 95-100 9530200-8 1998 Inhibition of the AP-1 transcription factor with curcumin decreased the cytokine induction of HO-1 mRNA, suggesting the involvement of this transcription factor in cytokine signaling of HO-1. Curcumin 49-57 heme oxygenase 1 Homo sapiens 94-98 9530200-8 1998 Inhibition of the AP-1 transcription factor with curcumin decreased the cytokine induction of HO-1 mRNA, suggesting the involvement of this transcription factor in cytokine signaling of HO-1. Curcumin 49-57 heme oxygenase 1 Homo sapiens 186-190 9514830-3 1998 In the substantia nigra of both PD and control specimens, moderate HO-1 immunoreactivity was consistently observed in neuromelanin-containing (dopaminergic) neurons. neuromelanin 118-130 heme oxygenase 1 Homo sapiens 67-71 9514830-9 1998 In addition, excessive cellular levels of heme-derived free iron and carbon monoxide resulting from HO-1 overactivity may contribute to the pathogenesis of PD. Heme 42-46 heme oxygenase 1 Homo sapiens 100-104 9514830-9 1998 In addition, excessive cellular levels of heme-derived free iron and carbon monoxide resulting from HO-1 overactivity may contribute to the pathogenesis of PD. Iron 60-64 heme oxygenase 1 Homo sapiens 100-104 9514830-9 1998 In addition, excessive cellular levels of heme-derived free iron and carbon monoxide resulting from HO-1 overactivity may contribute to the pathogenesis of PD. Carbon Monoxide 69-84 heme oxygenase 1 Homo sapiens 100-104 9486963-0 1998 Hemeoxygenase-1 inhibits human myometrial contractility via carbon monoxide and is upregulated by progesterone during pregnancy. Progesterone 98-110 heme oxygenase 1 Homo sapiens 0-15 9486963-6 1998 Reverse transcription-PCR analysis revealed that mRNA encoding HO-1 and HO-2 was undetected in explant cultures of nonlaboring pregnant myometrium under basal conditions, however, exposure to progesterone, but not estradiol-17beta, induced the expression of HO-1 and HO-2 mRNAs. Progesterone 192-204 heme oxygenase 1 Homo sapiens 63-76 9486963-6 1998 Reverse transcription-PCR analysis revealed that mRNA encoding HO-1 and HO-2 was undetected in explant cultures of nonlaboring pregnant myometrium under basal conditions, however, exposure to progesterone, but not estradiol-17beta, induced the expression of HO-1 and HO-2 mRNAs. Progesterone 192-204 heme oxygenase 1 Homo sapiens 258-271 9486963-7 1998 Progesterone also significantly induced HO-1 protein synthesis (n = 4, P < 0.001) while estradiol-17beta had no effect (n = 4). Progesterone 0-12 heme oxygenase 1 Homo sapiens 40-44 9337616-8 1997 Because either hemin alone or UVA radiation are able to lead to a refractoriness of the heme oxygenase-1 gene to reinduction by a second exposure to one or the other agent in human fibroblasts, we conclude that heme, or an as yet unidentified heme derivative, is involved in the refractoriness response. Hemin 15-20 heme oxygenase 1 Homo sapiens 88-104 9425252-2 1997 We have examined the effect of the NO donor sodium nitroprusside (SNP) on heme oxygenase-1 (HO-1) expression in human epidermal keratinocytes and investigated the contribution of the heme oxygenase pathway in the control of keratinocyte proliferation. Nitroprusside 44-64 heme oxygenase 1 Homo sapiens 74-90 9374724-0 1997 Regulation of heme oxygenase-1 gene expression in vascular smooth muscle cells by nitric oxide. Nitric Oxide 82-94 heme oxygenase 1 Homo sapiens 14-30 9374724-4 1997 We demonstrate that the NO donor spermine NONOate (SNN) increases steady-state levels of HO-1 mRNA in aortic vascular smooth muscle cells (aSMC) in both a time- and dose-dependent manner. spermine nitric oxide complex 33-49 heme oxygenase 1 Homo sapiens 89-93 9374724-4 1997 We demonstrate that the NO donor spermine NONOate (SNN) increases steady-state levels of HO-1 mRNA in aortic vascular smooth muscle cells (aSMC) in both a time- and dose-dependent manner. spermine nitric oxide complex 51-54 heme oxygenase 1 Homo sapiens 89-93 9374724-4 1997 We demonstrate that the NO donor spermine NONOate (SNN) increases steady-state levels of HO-1 mRNA in aortic vascular smooth muscle cells (aSMC) in both a time- and dose-dependent manner. asmc 139-143 heme oxygenase 1 Homo sapiens 89-93 9374724-6 1997 Inhibition of the NO-induced HO-1 mRNA expression by cycloheximide suggests that new protein synthesis is required for increased HO-1 gene expression. Cycloheximide 53-66 heme oxygenase 1 Homo sapiens 29-33 9374724-6 1997 Inhibition of the NO-induced HO-1 mRNA expression by cycloheximide suggests that new protein synthesis is required for increased HO-1 gene expression. Cycloheximide 53-66 heme oxygenase 1 Homo sapiens 129-133 9374724-8 1997 The antioxidant N-acetyl-L-cysteine markedly inhibited SNN-induced HO-1 mRNA expression, whereas peroxynitrite did not induce HO-1 expression in aSMC. Acetylcysteine 16-35 heme oxygenase 1 Homo sapiens 67-71 9374724-8 1997 The antioxidant N-acetyl-L-cysteine markedly inhibited SNN-induced HO-1 mRNA expression, whereas peroxynitrite did not induce HO-1 expression in aSMC. spermine nitric oxide complex 55-58 heme oxygenase 1 Homo sapiens 67-71 9407320-2 1998 Overexpression of HO-1 in endothelial cells (EC) might, therefore, protect against oxidative stress produced under these pathological conditions, by generation of CO, a vasodilator, and bilirubin, which has antioxidant properties that enhance blood vessel formation to counteract hypoxia-induced injury. Bilirubin 186-195 heme oxygenase 1 Homo sapiens 18-22 9434639-2 1997 We investigated cellular events leading to the heme oxygenase-1 gene expression induced by sublethal concentrations of glutathione depletors, phorone and diethyl maleate, in human fibroblastic cells. Glutathione 119-130 heme oxygenase 1 Homo sapiens 47-63 9434639-2 1997 We investigated cellular events leading to the heme oxygenase-1 gene expression induced by sublethal concentrations of glutathione depletors, phorone and diethyl maleate, in human fibroblastic cells. phorone 142-149 heme oxygenase 1 Homo sapiens 47-63 9434639-2 1997 We investigated cellular events leading to the heme oxygenase-1 gene expression induced by sublethal concentrations of glutathione depletors, phorone and diethyl maleate, in human fibroblastic cells. diethyl maleate 154-169 heme oxygenase 1 Homo sapiens 47-63 9434639-3 1997 Accumulation of heme oxygenase-1 mRNA by glutathione depletors was canceled by simultaneous treatment with cycloheximide, an inhibitor of protein synthesis; however, the inhibitory effect decreased when the inhibitor was added 30 min later. Glutathione 41-52 heme oxygenase 1 Homo sapiens 16-32 9434639-3 1997 Accumulation of heme oxygenase-1 mRNA by glutathione depletors was canceled by simultaneous treatment with cycloheximide, an inhibitor of protein synthesis; however, the inhibitory effect decreased when the inhibitor was added 30 min later. Cycloheximide 107-120 heme oxygenase 1 Homo sapiens 16-32 9434639-5 1997 Accumulation of heme oxygenase-1 and c-fos transcripts was abrogated in cells pretreated with 1,4-diazabicyclo[2.2.2]octane, an oxygen-free radical quencher. triethylenediamine 94-123 heme oxygenase 1 Homo sapiens 16-32 9434639-7 1997 Pretreatment of cells with PD 98059, an inhibitor of the extracellular-signal regulated kinase cascade, or the c-fos antisense oligodeoxynucleotide inhibited the heme oxygenase-1 induction elicited by glutathione depletion. Oligodeoxyribonucleotides 127-147 heme oxygenase 1 Homo sapiens 162-178 9434639-7 1997 Pretreatment of cells with PD 98059, an inhibitor of the extracellular-signal regulated kinase cascade, or the c-fos antisense oligodeoxynucleotide inhibited the heme oxygenase-1 induction elicited by glutathione depletion. Glutathione 201-212 heme oxygenase 1 Homo sapiens 162-178 9434639-8 1997 These observations indicated that c-Fos protein plays a role in heme oxygenase-1 gene expression induced by glutathione depletion-mediated oxidative stress in human fibroblasts. Glutathione 108-119 heme oxygenase 1 Homo sapiens 64-80 9402154-3 1997 Indirect immunofluorescence double labeling studies demonstrated a simultaneous increase of ICAM-1 and HO-1 after exposure of cells to heme for 24 hr. Heme 135-139 heme oxygenase 1 Homo sapiens 103-107 9402154-9 1997 These results suggest that upregulation of ICAM-1, VCAM-1, and E selectin expression is associated with oxidative stress induced by hemoglobin/heme and that HO-1 may play a modulating role via its ability to degrade heme to a substance with antioxidant properties. Heme 216-220 heme oxygenase 1 Homo sapiens 157-161 9337616-8 1997 Because either hemin alone or UVA radiation are able to lead to a refractoriness of the heme oxygenase-1 gene to reinduction by a second exposure to one or the other agent in human fibroblasts, we conclude that heme, or an as yet unidentified heme derivative, is involved in the refractoriness response. Heme 211-215 heme oxygenase 1 Homo sapiens 88-104 9380735-0 1997 Heme oxygenase 1 is required for mammalian iron reutilization. Iron 43-47 heme oxygenase 1 Homo sapiens 0-16 9276739-5 1997 Heme oxygenase-1 (HO-1), the inducible isoform of HO, was highly induced by mildly oxidized LDL, and augmented induction was observed with hemin pretreatment. Hemin 139-144 heme oxygenase 1 Homo sapiens 0-16 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Heme 37-41 heme oxygenase 1 Homo sapiens 55-60 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Biliverdine 103-113 heme oxygenase 1 Homo sapiens 37-53 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Biliverdine 103-113 heme oxygenase 1 Homo sapiens 55-60 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Carbon Monoxide 115-130 heme oxygenase 1 Homo sapiens 37-53 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Carbon Monoxide 115-130 heme oxygenase 1 Homo sapiens 55-60 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Iron 141-145 heme oxygenase 1 Homo sapiens 37-53 9380736-1 1997 Stressed mammalian cells up-regulate heme oxygenase 1 (Hmox1; EC 1.14.99.3), which catabolizes heme to biliverdin, carbon monoxide, and free iron. Iron 141-145 heme oxygenase 1 Homo sapiens 55-60 9276739-5 1997 Heme oxygenase-1 (HO-1), the inducible isoform of HO, was highly induced by mildly oxidized LDL, and augmented induction was observed with hemin pretreatment. Hemin 139-144 heme oxygenase 1 Homo sapiens 18-22 9276739-9 1997 Oxidized phospholipids in the mildly oxidized LDL appear to be responsible for HO-1 induction, since oxidized but not native arachidonic acid-containing phospholipids also induced HO-1. Phospholipids 9-22 heme oxygenase 1 Homo sapiens 79-83 9276739-9 1997 Oxidized phospholipids in the mildly oxidized LDL appear to be responsible for HO-1 induction, since oxidized but not native arachidonic acid-containing phospholipids also induced HO-1. Phospholipids 9-22 heme oxygenase 1 Homo sapiens 180-184 9276739-9 1997 Oxidized phospholipids in the mildly oxidized LDL appear to be responsible for HO-1 induction, since oxidized but not native arachidonic acid-containing phospholipids also induced HO-1. Phospholipids 153-166 heme oxygenase 1 Homo sapiens 180-184 9276739-10 1997 These results suggest that HO-1 induced by mildly oxidized LDL may protect against the induction of inflammatory responses in artery wall cells through the production of the antioxidants biliverdin and bilirubin. Biliverdine 187-197 heme oxygenase 1 Homo sapiens 27-31 9276739-10 1997 These results suggest that HO-1 induced by mildly oxidized LDL may protect against the induction of inflammatory responses in artery wall cells through the production of the antioxidants biliverdin and bilirubin. Bilirubin 202-211 heme oxygenase 1 Homo sapiens 27-31 9054378-1 1997 Conversion of heme to verdoheme by heme oxygenase-1 (HO-1) is thought to involve alpha-meso-hydroxylation and elimination of the meso-carbon as CO, a reaction supported by both H2O2 and NADPH-cytochrome P450 reductase/O2. Carbon 134-140 heme oxygenase 1 Homo sapiens 35-51 9295197-7 1997 Heme oxygenase-1 (HO-1) markedly reduced the activity of both the endogenous AD inhibitor and hemin, indicating that the endogenous inhibitor contains heme. Heme 151-155 heme oxygenase 1 Homo sapiens 0-16 9295197-7 1997 Heme oxygenase-1 (HO-1) markedly reduced the activity of both the endogenous AD inhibitor and hemin, indicating that the endogenous inhibitor contains heme. Heme 151-155 heme oxygenase 1 Homo sapiens 18-22 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Heme 130-134 heme oxygenase 1 Homo sapiens 88-92 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Heme 130-134 heme oxygenase 1 Homo sapiens 94-99 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Biliverdine 146-165 heme oxygenase 1 Homo sapiens 88-92 9266806-5 1997 In addition, at lower doses, acrolein caused induction of heme oxygenase 1 protein; however, stress protein 72 (SP72) was not induced. Acrolein 29-37 heme oxygenase 1 Homo sapiens 58-74 9225984-2 1997 Heme oxygenase-1 (HO-1), a key enzyme in heme catabolism, also functions as an antioxidant enzyme. Heme 41-45 heme oxygenase 1 Homo sapiens 0-16 9225984-2 1997 Heme oxygenase-1 (HO-1), a key enzyme in heme catabolism, also functions as an antioxidant enzyme. Heme 41-45 heme oxygenase 1 Homo sapiens 18-22 9354392-0 1997 Possible implications of the induction of human heme oxygenase-1 by nitric oxide donors. Nitric Oxide 68-80 heme oxygenase 1 Homo sapiens 48-64 9354392-1 1997 To explore the involvement of nitric oxide (NO) in the induction of heme oxygenase-1, an essential enzyme in heme catabolism, we studied the effects of NO donors on the expression of heme oxygenase-1 mRNA in HeLa human cervical cancer cells. Nitric Oxide 30-42 heme oxygenase 1 Homo sapiens 68-84 9354392-4 1997 We showed that sodium nitroprusside also increased the levels of heme oxygenase-1 protein. Nitroprusside 15-35 heme oxygenase 1 Homo sapiens 65-81 9354392-5 1997 The sodium nitroprusside-mediated increase in heme oxygenase-1 mRNA levels was abolished by treatment with actinomycin D. Nitroprusside 4-24 heme oxygenase 1 Homo sapiens 46-62 9354392-5 1997 The sodium nitroprusside-mediated increase in heme oxygenase-1 mRNA levels was abolished by treatment with actinomycin D. Dactinomycin 107-120 heme oxygenase 1 Homo sapiens 46-62 9190885-6 1997 Pretreatment with ebselen increased the expression of such stress proteins as heat shock protein 70 and heme oxygenase-1 (heat shock protein 32) in cardiac myocytes, as assessed by Western blotting. ebselen 18-25 heme oxygenase 1 Homo sapiens 104-120 9190885-7 1997 Expression of heat shock protein 70 was increased only at a higher dose of ebselen (30 microM), whereas expression of heme oxygenase-1 was markedly increased at a lower dose of ebselen (3 microM). ebselen 177-184 heme oxygenase 1 Homo sapiens 118-134 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Biliverdine 146-165 heme oxygenase 1 Homo sapiens 94-99 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Carbon Monoxide 167-169 heme oxygenase 1 Homo sapiens 88-92 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Carbon Monoxide 167-169 heme oxygenase 1 Homo sapiens 94-99 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Iron 174-176 heme oxygenase 1 Homo sapiens 88-92 9116047-1 1997 Heme oxygenase-2 (HO-2) is constitutively expressed in mammalian tissues; together with HO-1 (HSP32) it catalyzes the cleavage of heme to produce biliverdin IX alpha, CO and Fe. Iron 174-176 heme oxygenase 1 Homo sapiens 94-99 9054378-1 1997 Conversion of heme to verdoheme by heme oxygenase-1 (HO-1) is thought to involve alpha-meso-hydroxylation and elimination of the meso-carbon as CO, a reaction supported by both H2O2 and NADPH-cytochrome P450 reductase/O2. Hydrogen Peroxide 177-181 heme oxygenase 1 Homo sapiens 35-51 8816811-2 1996 Heme oxygenase-1 (HO-1) is inducible not only by its heme substrate, but also by a variety of agents causing oxidative stress. Heme 53-57 heme oxygenase 1 Homo sapiens 0-16 9596945-1 1997 OBJECTIVE: The purification and identification of heme oxygenase isoforms (HO-1) and HO-2 from human liver were described and Sn-protoporphyrin (SnPP) was used to inhibit HO-1 activity in order to provide a new method for prevention and treatment of neonatal jaundice. tin protoporphyrin IX 126-143 heme oxygenase 1 Homo sapiens 75-79 9596945-1 1997 OBJECTIVE: The purification and identification of heme oxygenase isoforms (HO-1) and HO-2 from human liver were described and Sn-protoporphyrin (SnPP) was used to inhibit HO-1 activity in order to provide a new method for prevention and treatment of neonatal jaundice. tin protoporphyrin IX 126-143 heme oxygenase 1 Homo sapiens 171-175 9596945-1 1997 OBJECTIVE: The purification and identification of heme oxygenase isoforms (HO-1) and HO-2 from human liver were described and Sn-protoporphyrin (SnPP) was used to inhibit HO-1 activity in order to provide a new method for prevention and treatment of neonatal jaundice. tin protoporphyrin IX 145-149 heme oxygenase 1 Homo sapiens 171-175 9596945-5 1997 The apparent molecular weight of HO-1 and HO-2 on SDS-PAGE was 30,000 and 36,000 under reducing conditions, respectively. Sodium Dodecyl Sulfate 50-53 heme oxygenase 1 Homo sapiens 33-46 9012361-15 1997 Higher levels of HO-1 may be a contributing factor for the undetectable levels of cytochrome P450 arachidonic acid metabolites, 20-HETE, in the adenocarcinoma. Arachidonic Acid 98-114 heme oxygenase 1 Homo sapiens 17-21 9073574-6 1997 As an example of such an approach, singlet oxygen has been implicated in the UVA radiation activation of HO-1 because the activation is enhanced by deuterium oxide (which enhances singlet oxygen lifetime) and suppressed by histidine (which scavenges the species). Singlet Oxygen 35-49 heme oxygenase 1 Homo sapiens 105-109 9073574-6 1997 As an example of such an approach, singlet oxygen has been implicated in the UVA radiation activation of HO-1 because the activation is enhanced by deuterium oxide (which enhances singlet oxygen lifetime) and suppressed by histidine (which scavenges the species). Deuterium Oxide 148-163 heme oxygenase 1 Homo sapiens 105-109 9073574-6 1997 As an example of such an approach, singlet oxygen has been implicated in the UVA radiation activation of HO-1 because the activation is enhanced by deuterium oxide (which enhances singlet oxygen lifetime) and suppressed by histidine (which scavenges the species). Singlet Oxygen 180-194 heme oxygenase 1 Homo sapiens 105-109 9073574-6 1997 As an example of such an approach, singlet oxygen has been implicated in the UVA radiation activation of HO-1 because the activation is enhanced by deuterium oxide (which enhances singlet oxygen lifetime) and suppressed by histidine (which scavenges the species). Histidine 223-232 heme oxygenase 1 Homo sapiens 105-109 8897897-3 1996 SNAP (300 microM) caused a transient increase in heme oxygenase-1 (HO-1) mRNA associated with a fivefold increase in HO activity that was completely blocked by the competitive HO inhibitor, tin protoporphyrin IX (SnPP). protoporphyrin IX 194-211 heme oxygenase 1 Homo sapiens 49-65 8897897-3 1996 SNAP (300 microM) caused a transient increase in heme oxygenase-1 (HO-1) mRNA associated with a fivefold increase in HO activity that was completely blocked by the competitive HO inhibitor, tin protoporphyrin IX (SnPP). S-Nitroso-N-propionyl-D,L-penicillamine 213-217 heme oxygenase 1 Homo sapiens 49-65 8816811-2 1996 Heme oxygenase-1 (HO-1) is inducible not only by its heme substrate, but also by a variety of agents causing oxidative stress. Heme 53-57 heme oxygenase 1 Homo sapiens 18-22 8816811-10 1996 Tin protoporphyrin, a selective inhibitor of HO, reversed the growth arrest and ablated the increased survival against hyperoxia observed in the A549-A4 cells overexpressing HO-1. tin protoporphyrin IX 0-18 heme oxygenase 1 Homo sapiens 174-178 8816811-11 1996 Taken together, our data suggest that overexpression of HO-1 results in cell growth arrest, which may facilitate cellular protection against non-heme-mediated oxidant insult such as hyperoxia. Heme 145-149 heme oxygenase 1 Homo sapiens 56-60 8783636-2 1996 BSC-1 cells adapted by long-term exposure to H2O2 exhibited a twofold increase in basal HO activity and expression of HO-1 mRNA as compared with their wild-type counterparts. Hydrogen Peroxide 45-49 heme oxygenase 1 Homo sapiens 118-122 8800205-3 1996 Among proteins induced after oxidative stress is a 32 kDa protein-probably corresponding to heme oxygenase-1 (HO-1)- and a 27 kDa protein, both known to be induced by reactive oxygen species. Reactive Oxygen Species 167-190 heme oxygenase 1 Homo sapiens 92-108 8800205-3 1996 Among proteins induced after oxidative stress is a 32 kDa protein-probably corresponding to heme oxygenase-1 (HO-1)- and a 27 kDa protein, both known to be induced by reactive oxygen species. Reactive Oxygen Species 167-190 heme oxygenase 1 Homo sapiens 110-114 8783636-3 1996 Exposure of both adapted and wild-type BSC-1 cells to H2O2 induced HO-1 mRNA. Hydrogen Peroxide 54-58 heme oxygenase 1 Homo sapiens 67-71 8783636-8 1996 In conclusion, two interrelated stressors, H2O2 and hemin, produced a stimulation of HO-1, and this was associated with a reduction in the viability of BSC-1 cells. Hydrogen Peroxide 43-47 heme oxygenase 1 Homo sapiens 85-89 8783636-8 1996 In conclusion, two interrelated stressors, H2O2 and hemin, produced a stimulation of HO-1, and this was associated with a reduction in the viability of BSC-1 cells. Hemin 52-57 heme oxygenase 1 Homo sapiens 85-89 8769112-0 1996 A precursor of the nitric oxide donor SIN-1 modulates the stress protein heme oxygenase-1 in rat liver. Nitric Oxide 19-31 heme oxygenase 1 Homo sapiens 73-89 8694803-1 1996 Heme oxygenase-1, a key enzyme in heme catabolism, and inducible nitric oxide synthase (iNOS) are responsible for production of carbon monoxide and nitric oxide (NO), respectively. Carbon Monoxide 128-143 heme oxygenase 1 Homo sapiens 0-16 8764571-0 1996 Expression of heme oxygenase isozyme mRNAs in the human brain and induction of heme oxygenase-1 by nitric oxide donors. Nitric Oxide 99-111 heme oxygenase 1 Homo sapiens 79-95 8764571-4 1996 In a human glioblastoma cell line, T98G, treatment with any of three types of NO donors--sodium nitroprusside, 3-morpholinosydnonimine, and S-nitroso-L-glutathione--caused a significant increase in the levels of heme oxygenase-1 mRNA but not in the levels of heme oxygenase-2 and heat-shock protein 70 mRNAs. Nitroprusside 89-109 heme oxygenase 1 Homo sapiens 212-228 8764571-4 1996 In a human glioblastoma cell line, T98G, treatment with any of three types of NO donors--sodium nitroprusside, 3-morpholinosydnonimine, and S-nitroso-L-glutathione--caused a significant increase in the levels of heme oxygenase-1 mRNA but not in the levels of heme oxygenase-2 and heat-shock protein 70 mRNAs. linsidomine 111-134 heme oxygenase 1 Homo sapiens 212-228 8764571-4 1996 In a human glioblastoma cell line, T98G, treatment with any of three types of NO donors--sodium nitroprusside, 3-morpholinosydnonimine, and S-nitroso-L-glutathione--caused a significant increase in the levels of heme oxygenase-1 mRNA but not in the levels of heme oxygenase-2 and heat-shock protein 70 mRNAs. S-Nitrosoglutathione 140-163 heme oxygenase 1 Homo sapiens 212-228 8764571-5 1996 Sodium nitroprusside increased the levels of heme oxygenase-1 protein but not the levels of heat-shock protein 70 in T98G cells. Nitroprusside 0-20 heme oxygenase 1 Homo sapiens 45-61 8764571-6 1996 The increase in content of heme oxygenase-1 mRNA caused by sodium nitro-prusside was completely abolished by the treatment with actinomycin D. Nitroprusside 59-80 heme oxygenase 1 Homo sapiens 27-43 8764571-6 1996 The increase in content of heme oxygenase-1 mRNA caused by sodium nitro-prusside was completely abolished by the treatment with actinomycin D. Dactinomycin 128-141 heme oxygenase 1 Homo sapiens 27-43 8735643-2 1996 In the feline lower oesophageal sphincter (LOS), the distribution of the carbon monoxide (CO) producing enzymes haem oxygenase (HO)-1 and -2 was studied by immunohistochemistry and confocal microscopy, the HO activity was measured and the possible role for CO as a mediator of relaxation was investigated. Carbon Monoxide 73-88 heme oxygenase 1 Homo sapiens 112-140 8679227-8 1996 Since the identification of HO-1 in 1968, many of the studies involving this enzyme were understandably focused on the regulation and function of HO-1 in heme metabolism. Heme 154-158 heme oxygenase 1 Homo sapiens 28-32 8679227-8 1996 Since the identification of HO-1 in 1968, many of the studies involving this enzyme were understandably focused on the regulation and function of HO-1 in heme metabolism. Heme 154-158 heme oxygenase 1 Homo sapiens 146-150 8679227-9 1996 This emphasis is self-evident as HO-1 catalyzes the first and rate-limiting step in heme degradation. Heme 84-88 heme oxygenase 1 Homo sapiens 33-37 8679227-10 1996 Interestingly, however, evidence accumulated over the past 25 years demonstrates that HO-1 is induced not only by the substrate heme but also by a variety of non-heme inducers such as heavy metals, endotoxin, heat shock, inflammatory cytokines, and prostaglandins. Heme 128-132 heme oxygenase 1 Homo sapiens 86-90 8679227-10 1996 Interestingly, however, evidence accumulated over the past 25 years demonstrates that HO-1 is induced not only by the substrate heme but also by a variety of non-heme inducers such as heavy metals, endotoxin, heat shock, inflammatory cytokines, and prostaglandins. Heme 162-166 heme oxygenase 1 Homo sapiens 86-90 8679227-10 1996 Interestingly, however, evidence accumulated over the past 25 years demonstrates that HO-1 is induced not only by the substrate heme but also by a variety of non-heme inducers such as heavy metals, endotoxin, heat shock, inflammatory cytokines, and prostaglandins. Prostaglandins 249-263 heme oxygenase 1 Homo sapiens 86-90 8679227-11 1996 The chemical diversity of HO-1 inducers led to the speculation that HO-1, besides its role in heme degradation, may also play a vital function in maintaining cellular homeostasis. Heme 94-98 heme oxygenase 1 Homo sapiens 26-30 8679227-11 1996 The chemical diversity of HO-1 inducers led to the speculation that HO-1, besides its role in heme degradation, may also play a vital function in maintaining cellular homeostasis. Heme 94-98 heme oxygenase 1 Homo sapiens 68-72 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Carbon Monoxide 195-210 heme oxygenase 1 Homo sapiens 17-21 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Carbon Monoxide 195-210 heme oxygenase 1 Homo sapiens 239-243 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Iron 212-216 heme oxygenase 1 Homo sapiens 17-21 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Iron 212-216 heme oxygenase 1 Homo sapiens 239-243 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Bilirubin 222-231 heme oxygenase 1 Homo sapiens 17-21 8679227-14 1996 The magnitude of HO-1 induction after oxidative stress and the wide distribution of this enzyme in systemic tissues coupled with the intriguing biological activities of the catalytic byproducts, carbon monoxide, iron, and bilirubin, makes HO-1 a highly attractive and interesting candidate stress-response protein which may play key role(s) in mediating protection against oxidant-mediated lung injury. Bilirubin 222-231 heme oxygenase 1 Homo sapiens 239-243 8652820-7 1996 Interestingly, heat shock abolished the remarkable increase in the levels of heme oxygenase-1 mRNA in YN-1-0-A cells treated with hemin or cadmium, in which HSP70 mRNA was noticeably induced. Hemin 130-135 heme oxygenase 1 Homo sapiens 77-93 8652820-7 1996 Interestingly, heat shock abolished the remarkable increase in the levels of heme oxygenase-1 mRNA in YN-1-0-A cells treated with hemin or cadmium, in which HSP70 mRNA was noticeably induced. Cadmium 139-146 heme oxygenase 1 Homo sapiens 77-93 8652820-8 1996 Furthermore, transient expression assays showed that heat shock inhibits the cadmium-mediated activation of the heme oxygenase-1 promoter, whereas the HSP70 gene promoter was activated upon heat shock. Cadmium 77-84 heme oxygenase 1 Homo sapiens 112-128 8735643-2 1996 In the feline lower oesophageal sphincter (LOS), the distribution of the carbon monoxide (CO) producing enzymes haem oxygenase (HO)-1 and -2 was studied by immunohistochemistry and confocal microscopy, the HO activity was measured and the possible role for CO as a mediator of relaxation was investigated. Carbon Monoxide 90-92 heme oxygenase 1 Homo sapiens 112-140 8675634-1 1995 CO is produced in vascular smooth muscle cells (VSMC) by heme oxygenase-1 (HO-1). Carbon Monoxide 0-2 heme oxygenase 1 Homo sapiens 57-73 8650873-1 1996 OBJECTIVES: Heme oxygenase isozymes, HO-1 and HO-2, are members of the stress/heat shock (HSP) family of proteins, with the known function of cleaving the heme molecule to biliverdin, iron, and carbon monoxide. Heme 155-159 heme oxygenase 1 Homo sapiens 37-50 8650873-1 1996 OBJECTIVES: Heme oxygenase isozymes, HO-1 and HO-2, are members of the stress/heat shock (HSP) family of proteins, with the known function of cleaving the heme molecule to biliverdin, iron, and carbon monoxide. Biliverdine 172-182 heme oxygenase 1 Homo sapiens 37-50 8650873-1 1996 OBJECTIVES: Heme oxygenase isozymes, HO-1 and HO-2, are members of the stress/heat shock (HSP) family of proteins, with the known function of cleaving the heme molecule to biliverdin, iron, and carbon monoxide. Iron 184-188 heme oxygenase 1 Homo sapiens 37-50 8650873-1 1996 OBJECTIVES: Heme oxygenase isozymes, HO-1 and HO-2, are members of the stress/heat shock (HSP) family of proteins, with the known function of cleaving the heme molecule to biliverdin, iron, and carbon monoxide. Carbon Monoxide 194-209 heme oxygenase 1 Homo sapiens 37-50 8650873-8 1996 HO-1 in the prostate tissue was found catalytically active and oxidatively cleaved the heme molecule (Fe-protoporphyrin IX) to biliverdin. Heme 87-91 heme oxygenase 1 Homo sapiens 0-4 8650873-8 1996 HO-1 in the prostate tissue was found catalytically active and oxidatively cleaved the heme molecule (Fe-protoporphyrin IX) to biliverdin. ferriheme b(1-) 102-122 heme oxygenase 1 Homo sapiens 0-4 8650873-8 1996 HO-1 in the prostate tissue was found catalytically active and oxidatively cleaved the heme molecule (Fe-protoporphyrin IX) to biliverdin. Biliverdine 127-137 heme oxygenase 1 Homo sapiens 0-4 8650873-12 1996 CONCLUSIONS: The finding that HO-1 expression is increased in BPH and malignant prostate tissue is consistent with a role for this stress protein in the pathogenesis of BPH and prostate cancer; in the context of iron metabolism, an argument is made in support of this possibility. Iron 212-216 heme oxygenase 1 Homo sapiens 30-34 8547275-0 1996 Heme oxygenase (HO-1): His-132 stabilizes a distal water ligand and assists catalysis. Histidine 23-26 heme oxygenase 1 Homo sapiens 16-20 8547275-0 1996 Heme oxygenase (HO-1): His-132 stabilizes a distal water ligand and assists catalysis. Water 51-56 heme oxygenase 1 Homo sapiens 16-20 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Histidine 0-3 heme oxygenase 1 Homo sapiens 83-107 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Histidine 0-3 heme oxygenase 1 Homo sapiens 109-113 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Histidine 11-14 heme oxygenase 1 Homo sapiens 83-107 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Histidine 11-14 heme oxygenase 1 Homo sapiens 109-113 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Heme 63-67 heme oxygenase 1 Homo sapiens 83-107 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Heme 63-67 heme oxygenase 1 Homo sapiens 109-113 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Iron 68-72 heme oxygenase 1 Homo sapiens 83-107 8547275-1 1996 His-25 and His-132 are the primary candidates for the proximal heme iron ligand in heme oxygenase isozyme-1 (HO-1). Iron 68-72 heme oxygenase 1 Homo sapiens 109-113 8547275-7 1996 These results place His-132 close to the iron on the distal side of the heme pocket and indicate that His-132 facilitates, but is not absolutely required for, the catalytic turnover of HO-1. Histidine 20-23 heme oxygenase 1 Homo sapiens 185-189 8547275-7 1996 These results place His-132 close to the iron on the distal side of the heme pocket and indicate that His-132 facilitates, but is not absolutely required for, the catalytic turnover of HO-1. Heme 72-76 heme oxygenase 1 Homo sapiens 185-189 8547275-7 1996 These results place His-132 close to the iron on the distal side of the heme pocket and indicate that His-132 facilitates, but is not absolutely required for, the catalytic turnover of HO-1. Histidine 102-105 heme oxygenase 1 Homo sapiens 185-189 8654390-1 1996 Heme oxygenase 1 is an essential enzyme in heme catabolism that cleaves heme to form biliverdin, iron, and carbon monoxide. Heme 43-47 heme oxygenase 1 Homo sapiens 0-16 8654390-1 1996 Heme oxygenase 1 is an essential enzyme in heme catabolism that cleaves heme to form biliverdin, iron, and carbon monoxide. Heme 72-76 heme oxygenase 1 Homo sapiens 0-16 8654390-1 1996 Heme oxygenase 1 is an essential enzyme in heme catabolism that cleaves heme to form biliverdin, iron, and carbon monoxide. Biliverdine 85-95 heme oxygenase 1 Homo sapiens 0-16 8654390-1 1996 Heme oxygenase 1 is an essential enzyme in heme catabolism that cleaves heme to form biliverdin, iron, and carbon monoxide. Iron 97-101 heme oxygenase 1 Homo sapiens 0-16 8654390-1 1996 Heme oxygenase 1 is an essential enzyme in heme catabolism that cleaves heme to form biliverdin, iron, and carbon monoxide. Carbon Monoxide 107-122 heme oxygenase 1 Homo sapiens 0-16 8654390-2 1996 The human heme-oxygenase-1 gene is transcriptionally activated through the cis-regulatory element (MTE), GTCATATGAC (positions -156 to -147), during 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced differentiation of myelomonocytic cell lines, such as THP-1, to macrophages. Tetradecanoylphorbol Acetate 149-185 heme oxygenase 1 Homo sapiens 10-26 8654390-2 1996 The human heme-oxygenase-1 gene is transcriptionally activated through the cis-regulatory element (MTE), GTCATATGAC (positions -156 to -147), during 12-O-tetradecanoylphorbol 13-acetate (TPA)-induced differentiation of myelomonocytic cell lines, such as THP-1, to macrophages. Tetradecanoylphorbol Acetate 187-190 heme oxygenase 1 Homo sapiens 10-26 8743975-6 1996 These results indicate that peroxidation of internal membrane lipids, a decrease in the intracellular GSH levels and the integrity of the plasma membrane are all important for the UVA-induction of heme oxygenase-1. Glutathione 102-105 heme oxygenase 1 Homo sapiens 197-213 8743975-8 1996 The nonenzymatic lipid peroxidation product 4-hydroxynonenal induces heme oxygenase-1 mRNA up to 40-fold and the phospholipase metabolites diacylglycerol and arachidonic acid induce this mRNA by three-to sixfold above basal levels. 4-hydroxy-2-nonenal 44-60 heme oxygenase 1 Homo sapiens 69-85 8743975-9 1996 We also demonstrate that the cyclooxygenase metabolites of arachidonic acid are important for the UVA-activation of the heme oxygenase-1 gene. Arachidonic Acid 59-75 heme oxygenase 1 Homo sapiens 120-136 8600294-8 1995 Hyperthermia and treatment with Na arsenite both resulted in enhanced expression of HSP70 and HO-1. na arsenite 32-43 heme oxygenase 1 Homo sapiens 94-98 8600294-9 1995 In addition, exposure to hydrogen peroxide (H2O2), cadmium (Cd), and lipopolysaccharide (LPS) stimulated a rise in HO-1 levels. Hydrogen Peroxide 25-42 heme oxygenase 1 Homo sapiens 115-119 8600294-9 1995 In addition, exposure to hydrogen peroxide (H2O2), cadmium (Cd), and lipopolysaccharide (LPS) stimulated a rise in HO-1 levels. Hydrogen Peroxide 44-48 heme oxygenase 1 Homo sapiens 115-119 8600294-9 1995 In addition, exposure to hydrogen peroxide (H2O2), cadmium (Cd), and lipopolysaccharide (LPS) stimulated a rise in HO-1 levels. Cadmium 51-58 heme oxygenase 1 Homo sapiens 115-119 8600294-9 1995 In addition, exposure to hydrogen peroxide (H2O2), cadmium (Cd), and lipopolysaccharide (LPS) stimulated a rise in HO-1 levels. Cadmium 60-62 heme oxygenase 1 Homo sapiens 115-119 7476027-3 1995 In the present study, we determined that CSH treatment resulted in an increase in HSP 27, HSP 90 and heme oxygenase (HO-1) at both the protein and mRNA level. csh 41-44 heme oxygenase 1 Homo sapiens 117-121 7545388-8 1995 Induction of heme oxygenase-1 and its inhibition by Sn protoporphyrin IX had no effect on cGMP levels. Tin 52-54 heme oxygenase 1 Homo sapiens 13-29