PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 15950210-11 2005 Thus, 9,10-PQ inhibits GAPDH by two distinct mechanisms: through ROS generation that results in the oxidization of GAPDH thiols, and by an oxygen-independent mechanism that results in the modification of GAPDH catalytic thiols. 9,10-phenanthrenequinone 6-13 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 23-28 17847711-2 2007 9,10-Phenanthrenequinone (9,10-PQ) and 1,2-naphthoquinone (1,2-NQ), which are contained in diesel exhaust particles (DEPs), potently inhibited 20alpha-HSD activity in liver cytosol. 9,10-phenanthrenequinone 0-24 aldo-keto reductase family 1, member C18 Mus musculus 143-154 17847711-2 2007 9,10-Phenanthrenequinone (9,10-PQ) and 1,2-naphthoquinone (1,2-NQ), which are contained in diesel exhaust particles (DEPs), potently inhibited 20alpha-HSD activity in liver cytosol. 9,10-phenanthrenequinone 26-33 aldo-keto reductase family 1, member C18 Mus musculus 143-154 17082565-6 2007 We found that both acenaphthenequinone (AcQ) and 9,10-phenanthrenequinone (PQ) enhanced ROS generation and that AcQ translocated NF-kappaB from the cytosol to the nucleus. 9,10-phenanthrenequinone 75-77 nuclear factor kappa B subunit 1 Homo sapiens 129-138 16164454-9 2005 PQ exhibited adjuvant activity for the allergen-specific production of IgG1 and IgE. 9,10-phenanthrenequinone 0-2 LOC105243590 Mus musculus 71-75 15950210-0 2005 The interactions of 9,10-phenanthrenequinone with glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a potential site for toxic actions. 9,10-phenanthrenequinone 20-44 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 50-90 15950210-0 2005 The interactions of 9,10-phenanthrenequinone with glyceraldehyde-3-phosphate dehydrogenase (GAPDH), a potential site for toxic actions. 9,10-phenanthrenequinone 20-44 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 92-97 15950210-3 2005 As part of a continuing study of the interactions of quinones with biological systems, we have examined the susceptibility of GAPDH to inactivation by 9,10-phenanthrenequinone (9,10-PQ). 9,10-phenanthrenequinone 151-175 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 126-131 15950210-3 2005 As part of a continuing study of the interactions of quinones with biological systems, we have examined the susceptibility of GAPDH to inactivation by 9,10-phenanthrenequinone (9,10-PQ). 9,10-phenanthrenequinone 177-184 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 126-131 15950210-5 2005 In this study, the effects of 9,10-PQ on GAPDH were examined in detail under aerobic and anaerobic conditions so that the role of oxygen could be distinguished from the direct effects of the quinone. 9,10-phenanthrenequinone 30-37 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 41-46 15950210-6 2005 The results indicate that, in the presence of the reducing agent DTT, GAPDH inhibition by 9,10-PQ under aerobic conditions was mostly indirect and comparable to the direct actions of exogenously-added H2O2 on this enzyme. 9,10-phenanthrenequinone 90-97 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 70-75 15950210-7 2005 GAPDH was also inhibited by 9,10-PQ anaerobically, but in a somewhat more complex manner. 9,10-phenanthrenequinone 28-35 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 0-5 15950210-11 2005 Thus, 9,10-PQ inhibits GAPDH by two distinct mechanisms: through ROS generation that results in the oxidization of GAPDH thiols, and by an oxygen-independent mechanism that results in the modification of GAPDH catalytic thiols. 9,10-phenanthrenequinone 6-13 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 115-120 15950210-11 2005 Thus, 9,10-PQ inhibits GAPDH by two distinct mechanisms: through ROS generation that results in the oxidization of GAPDH thiols, and by an oxygen-independent mechanism that results in the modification of GAPDH catalytic thiols. 9,10-phenanthrenequinone 6-13 glyceraldehyde-3-phosphate dehydrogenase Homo sapiens 115-120 12604216-8 2003 Bacterially expressed recombinant HTSP protein showed small but significant activity towards 9,10-phenanthrenequinone among the putative substrates so far tested. 9,10-phenanthrenequinone 93-117 aldo-keto reductase family 1 member E2 Homo sapiens 34-38 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 superoxide dismutase 1 Homo sapiens 65-74 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 15629867-1 2005 9,10-Phenanthraquinone (PQ), a major quinone contained in diesel exhaust particles and atmospheric PM(2.5), undergoes one-electron reduction by flavin enzymes such as NADPH-cytochrome P450 reductase, leading to production of reactive oxygen species in vitro. 9,10-phenanthrenequinone 0-22 cytochrome p450 oxidoreductase Homo sapiens 167-198 15629867-1 2005 9,10-Phenanthraquinone (PQ), a major quinone contained in diesel exhaust particles and atmospheric PM(2.5), undergoes one-electron reduction by flavin enzymes such as NADPH-cytochrome P450 reductase, leading to production of reactive oxygen species in vitro. 9,10-phenanthrenequinone 24-26 cytochrome p450 oxidoreductase Homo sapiens 167-198 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 superoxide dismutase 1 Homo sapiens 113-139 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 superoxide dismutase 1 Homo sapiens 141-150 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 15629867-9 2005 The inhibitor of Cu,Zn-SOD, diethyldithiocarbamate, enhanced the toxic effects of 5 microM PQ. 9,10-phenanthrenequinone 91-93 superoxide dismutase 1 Homo sapiens 17-26 15629867-10 2005 The present findings suggest that PQ causes iron-mediated oxidative damage that is exacerbated by the concomitant down-regulation of Cu,Zn-SOD. 9,10-phenanthrenequinone 34-36 superoxide dismutase 1 Homo sapiens 133-142 15669044-8 2005 Phenanthraquinone induced the lung expression of interleukin (IL)-5 and eotaxin 48 h and 24 h after the challenge, respectively. 9,10-phenanthrenequinone 0-17 interleukin 5 Mus musculus 49-67 15669044-8 2005 Phenanthraquinone induced the lung expression of interleukin (IL)-5 and eotaxin 48 h and 24 h after the challenge, respectively. 9,10-phenanthrenequinone 0-17 chemokine (C-C motif) ligand 11 Mus musculus 72-79 15669044-9 2005 These results indicate that intratracheal exposure to phenanthraquinone induces recruitment of inflammatory cells, at least partly, through the local expression of IL-5 and eotaxin. 9,10-phenanthrenequinone 54-71 interleukin 5 Mus musculus 164-168 15669044-9 2005 These results indicate that intratracheal exposure to phenanthraquinone induces recruitment of inflammatory cells, at least partly, through the local expression of IL-5 and eotaxin. 9,10-phenanthrenequinone 54-71 chemokine (C-C motif) ligand 11 Mus musculus 173-180 15310246-2 2004 PHCR has the ability to catalyze efficiently the reduction of 9,10-phenanthrenequinone (PQ) contained in diesel exhaust particles (DEPs). 9,10-phenanthrenequinone 62-86 dehydrogenase/reductase SDR family member 4 Sus scrofa 0-4 15310246-2 2004 PHCR has the ability to catalyze efficiently the reduction of 9,10-phenanthrenequinone (PQ) contained in diesel exhaust particles (DEPs). 9,10-phenanthrenequinone 88-90 dehydrogenase/reductase SDR family member 4 Sus scrofa 0-4 15310246-6 2004 These results indicate that PQ inhibits the reduction of 4-BP and all-trans-retinal by acting PHCR present in pig heart cytosol. 9,10-phenanthrenequinone 28-30 dehydrogenase/reductase SDR family member 4 Sus scrofa 94-98 15310246-8 2004 The absorbance of cytochrome c at 550 nm was increased with the time by adding PQ, and the increased absorbance was decreased in the presence of superoxide dismutase. 9,10-phenanthrenequinone 79-81 cytochrome c Sus scrofa 18-30 15310246-10 2004 On the basis of these results, it is concluded that PQ not only inhibits the reduction of 4-BP and all-trans-retinal catalyzed by PHCR but also mediates superoxide formation through its redox cycling involved in PHCR. 9,10-phenanthrenequinone 52-54 dehydrogenase/reductase SDR family member 4 Sus scrofa 130-134 15310246-10 2004 On the basis of these results, it is concluded that PQ not only inhibits the reduction of 4-BP and all-trans-retinal catalyzed by PHCR but also mediates superoxide formation through its redox cycling involved in PHCR. 9,10-phenanthrenequinone 52-54 dehydrogenase/reductase SDR family member 4 Sus scrofa 212-216 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 263-280 nitric oxide synthase 3 Rattus norvegicus 290-305 11404275-0 2001 Phenanthraquinone inhibits eNOS activity and suppresses vasorelaxation. 9,10-phenanthrenequinone 0-17 nitric oxide synthase 3 Rattus norvegicus 27-31 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 263-280 nitric oxide synthase 3 Rattus norvegicus 307-311 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 1 Rattus norvegicus 129-159 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 1 Rattus norvegicus 161-165 11404275-8 2001 The present findings suggest that phenanthraquinone has a potent inhibitory action on eNOS activity via a similar mechanism reported for nNOS, thereby causing the suppression of NO-mediated vasorelaxation and elevation of blood pressure. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 3 Rattus norvegicus 86-90 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 3 Rattus norvegicus 290-305 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 3 Rattus norvegicus 307-311 11404275-8 2001 The present findings suggest that phenanthraquinone has a potent inhibitory action on eNOS activity via a similar mechanism reported for nNOS, thereby causing the suppression of NO-mediated vasorelaxation and elevation of blood pressure. 9,10-phenanthrenequinone 34-51 nitric oxide synthase 1 Rattus norvegicus 137-141 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 263-280 nitric oxide synthase 1 Rattus norvegicus 129-159 11356112-3 2001 A number of 9,10-phenanthrenediones were identified that reversibly inhibited CD45-mediated p-nitrophenyl phosphate (pNPP) hydrolysis. 9,10-phenanthrenequinone 12-35 protein tyrosine phosphatase receptor type C Homo sapiens 78-82 11404275-2 2001 Since we reported previously that phenanthraquinone, an environmental chemical contained in diesel exhaust particles, suppresses neuronal nitric oxide synthase (nNOS) activity by shunting electrons away from the normal catalytic pathway, it was hypothesized that phenanthraquinone inhibits endothelial NOS (eNOS) activity and affects vascular tone. 9,10-phenanthrenequinone 263-280 nitric oxide synthase 1 Rattus norvegicus 161-165 9692994-4 1998 Surprisingly, Y55F and Y55S mutants of 3alpha-HSD reduced 9,10-PQ with high kcat values. 9,10-phenanthrenequinone 58-65 aldo-keto reductase family 1 member C4 Homo sapiens 39-49 10510318-4 1999 Also, AKR7A2 was found to exhibit a narrow substrate specificity, with activity being restricted to succinic semialdehyde (SSA), 2-nitrobenzaldehyde, pyridine-2-aldehyde, isatin, 1,2-naphthoquinone (1,2-NQ) and 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 211-235 aldo-keto reductase family 7 member A2 Homo sapiens 6-12 10622721-2 1999 The human PGFS catalyzed the reduction of prostaglandin (PG) D2, PGH2 and phenanthrenequinone (PQ), and the oxidation of 9alpha,11beta-PGF2 to PGD2. 9,10-phenanthrenequinone 74-93 aldo-keto reductase family 1 member C3 Homo sapiens 10-14 10622721-2 1999 The human PGFS catalyzed the reduction of prostaglandin (PG) D2, PGH2 and phenanthrenequinone (PQ), and the oxidation of 9alpha,11beta-PGF2 to PGD2. 9,10-phenanthrenequinone 95-97 aldo-keto reductase family 1 member C3 Homo sapiens 10-14 9625728-5 1998 Partial purification of the enzymes which are responsible for reducing PQ in 20000g supernatant of rat cerebellum by anion-exchange column chromatography indicated that one catalyst for PQ reduction was nNOS. 9,10-phenanthrenequinone 71-73 nitric oxide synthase 1 Rattus norvegicus 203-207 9625728-7 1998 nNOS effectively reduced the quinones as well as PQ causing a marked decrease in the production of NO from l-arginine, while 1, 4-benzoquinone, 9,10-anthraquinone, mitomycin C, and lapachol, which show negligible inhibitory action on nNOS activity, were poor substrates for the enzyme on reduction. 9,10-phenanthrenequinone 49-51 nitric oxide synthase 1 Rattus norvegicus 0-4 9625728-5 1998 Partial purification of the enzymes which are responsible for reducing PQ in 20000g supernatant of rat cerebellum by anion-exchange column chromatography indicated that one catalyst for PQ reduction was nNOS. 9,10-phenanthrenequinone 186-188 nitric oxide synthase 1 Rattus norvegicus 203-207 9625728-7 1998 nNOS effectively reduced the quinones as well as PQ causing a marked decrease in the production of NO from l-arginine, while 1, 4-benzoquinone, 9,10-anthraquinone, mitomycin C, and lapachol, which show negligible inhibitory action on nNOS activity, were poor substrates for the enzyme on reduction. 9,10-phenanthrenequinone 49-51 nitric oxide synthase 1 Rattus norvegicus 234-238 9625728-8 1998 These results indicate that PQ and other quinones used in the present study interact with the NADPH-cytochrome P450 reductase domain on nNOS and thus probably inhibit NO formation by shunting electrons away from the normal catalytic pathway. 9,10-phenanthrenequinone 28-30 nitric oxide synthase 1 Rattus norvegicus 136-140 9625728-6 1998 Reductase activity of PQ by purified nNOS required CaCl2/calmodulin and was markedly suppressed by the flavoprotein inhibitor diphenyleneiodonium but not by l-nitroarginine which is a specific inhibitor for NO formation. 9,10-phenanthrenequinone 22-24 nitric oxide synthase 1 Rattus norvegicus 37-41 9625728-6 1998 Reductase activity of PQ by purified nNOS required CaCl2/calmodulin and was markedly suppressed by the flavoprotein inhibitor diphenyleneiodonium but not by l-nitroarginine which is a specific inhibitor for NO formation. 9,10-phenanthrenequinone 22-24 calmodulin 1 Rattus norvegicus 57-67 8665510-5 1996 The immunodepletion experiments also revealed the presence of at least one other inducible carbonyl-reducing enzyme that, like AFAR, can metabolize 9,10-phenanthraquinone. 9,10-phenanthrenequinone 148-170 aldo-keto reductase family 7 member A3 Rattus norvegicus 127-131 1378964-3 1992 Synthesized GAA was separated by HPLC and detected fluorometrically after reacting with 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 88-112 alpha glucosidase Rattus norvegicus 12-15 8579568-1 1996 Guinea pig lens zeta-crystallin showed hyperbolic saturation curves with 9,10-phenanthrenequinone (PAQ). 9,10-phenanthrenequinone 73-97 quinone oxidoreductase Cavia porcellus 16-31 8579568-1 1996 Guinea pig lens zeta-crystallin showed hyperbolic saturation curves with 9,10-phenanthrenequinone (PAQ). 9,10-phenanthrenequinone 99-102 quinone oxidoreductase Cavia porcellus 16-31 8579568-3 1996 Whereas camel lens zeta-crystallin showed hyperbolic saturation curves only with PAQ and NADPH, but slightly segmoidal with juglone. 9,10-phenanthrenequinone 81-84 quinone oxidoreductase Cavia porcellus 19-34 8579568-5 1996 The catalytic center activity (Kcat) values indicated that camel zeta-crystallin catalyzed the reduction of PAQ more efficiently than the guinea pig lens zeta-crystallin, although the Km values of the two enzymes for this quinone were very similar. 9,10-phenanthrenequinone 108-111 quinone oxidoreductase Cavia porcellus 65-80 7811392-6 1994 A number of phenanthrene derivatives having a 9-hydroxy or 9-keto substituent, namely phenanthrenequinone, 6(5H)-phenanthridinone and 9-phenanthrol are potent inhibitors of bovine heart cAK (IC50 values 8, 10 and 10 microM, respectively) and of MLCK (IC50 values 6, 53 and 10 microM, respectively). 9,10-phenanthrenequinone 86-105 myosin light chain kinase, smooth muscle Bos taurus 245-249 8615688-4 1996 Since 19 alpha-estradiol is a competitive inhibitor of 9,10-phenanthrenequinone by the 17beta-hydroxysteroid dehydrogenase, it is likely that both reactions occur at the same active site on the enzyme. 9,10-phenanthrenequinone 55-79 hydroxysteroid 17-beta dehydrogenase 7 Homo sapiens 87-122 8526867-2 1995 This enzyme, designated AFAR, displays high activity towards dicarbonyl-containing compounds with ketone groups on adjacent carbon atoms; 9,10-phenanthrenequinone, acenaphthenequinone and camphorquinone were found to be good substrates. 9,10-phenanthrenequinone 138-162 aldo-keto reductase family 7 member A3 Rattus norvegicus 24-28 8526867-5 1995 Determination of the apparent Km reveals that AFAR has highest affinity for 9,10-phenanthrenequinone and succinic semialdehyde, and low affinity for glyoxal and DL-glyceraldehyde. 9,10-phenanthrenequinone 76-100 aldo-keto reductase family 7 member A3 Rattus norvegicus 46-50 7515872-3 1994 Transfection of the 3 alpha-HSD cDNA in Simian COS7 cells resulted in the expression of an immunoreactive protein to the antibodies against the purified enzyme, and the transfected cells exhibited activities for not only 7 alpha-hydroxy-5 beta-cholestan-3-one, the intermediate of bile acid biosynthesis, but also steroid hormones and 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 335-359 aldo-keto reductase family 1, member C14 Rattus norvegicus 20-31 7685581-3 1993 In a buffer containing 1% albumin and 10 microM quinone, 9,10-phenanthrenequinone is reduced most rapidly by the carbonyl reductase, 2-methyl-1,4-naphthoquinone is reduced most rapidly by the rat enzyme, and 3,6-pyrenequinone is reduced most rapidly by the Clostridium enzyme. 9,10-phenanthrenequinone 57-81 dehydrogenase/reductase 4 Rattus norvegicus 113-131 34391839-0 2021 9,10-Phenanthrenequinone provokes dysfunction of brain endothelial barrier through down-regulating expression of claudin-5. 9,10-phenanthrenequinone 0-24 claudin 5 Homo sapiens 113-122 34391839-5 2021 Immunofluorescence observation and Western blotting analysis also revealed that the 9,10-PQ treatment remarkably down-regulated the intercellular localization and expression of claudin-5 (CLDN5), a tight junctional protein that plays a key role in function of the blood-brain barrier, and the down-regulation was markedly recovered by pretreatment with a proteasome inhibitor Z-Leu-Leu-Leu-CHO. 9,10-phenanthrenequinone 84-91 claudin 5 Homo sapiens 177-186 34391839-5 2021 Immunofluorescence observation and Western blotting analysis also revealed that the 9,10-PQ treatment remarkably down-regulated the intercellular localization and expression of claudin-5 (CLDN5), a tight junctional protein that plays a key role in function of the blood-brain barrier, and the down-regulation was markedly recovered by pretreatment with a proteasome inhibitor Z-Leu-Leu-Leu-CHO. 9,10-phenanthrenequinone 84-91 claudin 5 Homo sapiens 188-193 34391839-6 2021 This result may indicate that sublethal concentrations of 9,10-PQ facilitate the dysfunction of the endothelial cell barrier through lowering in the expression and proteasomal proteolysis of CLDN5. 9,10-phenanthrenequinone 58-65 claudin 5 Homo sapiens 191-196 34391839-7 2021 The treatment with 9,10-PQ promoted nitric oxide (NO) production presumably through the induction of inducible NO synthase. 9,10-phenanthrenequinone 19-26 nitric oxide synthase 2 Homo sapiens 101-122 34391839-8 2021 In addition, the 9,10-PQ-mediated down-regulation of CLDN5 was ameliorated and deteriorated by pretreating with a scavenger and donor, respectively, of NO. 9,10-phenanthrenequinone 17-24 claudin 5 Homo sapiens 53-58 34391839-9 2021 Similarly to the 9,10-PQ treatment, treatment with a donor of peroxynitrite, a highly reactive oxidant formed by the reaction of NO and superoxide anion, resulted in the marked reduction of CLDN5 expression and elevation of 26S proteasome-based proteolytic activities. 9,10-phenanthrenequinone 17-24 claudin 5 Homo sapiens 190-195 7108550-1 1982 Guanidino compounds in CSF of 57 human subjects were determined fluorometrically after reaction with phenanthrenequinone in alkali solution, using HPLC. 9,10-phenanthrenequinone 101-120 colony stimulating factor 2 Homo sapiens 23-26 6584903-6 1984 For example, phenanthrenequinone was converted to a nonmutagenic metabolite in a cytochrome P-450-dependent reaction, whereas danthron was converted to a highly mutagenic metabolite. 9,10-phenanthrenequinone 13-32 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 81-97 2910843-12 1989 The PGH2 9,11-endoperoxide reductase activity of aldehyde reductase was not affected in the presence of a substrate such as p-nitrobenzaldehyde, DL-glyceraldehyde, or 9,10-phenanthrenequinone, suggesting that PGH2 9,11-endoperoxide and other substrates are reduced at different active site(s). 9,10-phenanthrenequinone 167-191 aldo-keto reductase family 1 member A1 Homo sapiens 49-67 33483563-6 2021 Substrate screening suggested that VAT-1 possesses oxidoreductase activity against quinones such as 1,2-naphthoquinone and 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 123-147 vesicle amine transport 1 Homo sapiens 35-40 33483563-6 2021 Substrate screening suggested that VAT-1 possesses oxidoreductase activity against quinones such as 1,2-naphthoquinone and 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 123-147 thioredoxin reductase 1 Homo sapiens 51-65 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 181-188 epidermal growth factor receptor Homo sapiens 156-160 32493877-0 2020 Redox cycling of 9,10-phenanthrenequinone activates epidermal growth factor receptor signaling through S-oxidation of protein tyrosine phosphatase 1B. 9,10-phenanthrenequinone 17-41 epidermal growth factor receptor Homo sapiens 52-84 32493877-0 2020 Redox cycling of 9,10-phenanthrenequinone activates epidermal growth factor receptor signaling through S-oxidation of protein tyrosine phosphatase 1B. 9,10-phenanthrenequinone 17-41 protein tyrosine phosphatase non-receptor type 1 Homo sapiens 118-149 32493877-3 2020 The current study examined whether 9,10-PQ could activate epidermal growth factor receptor (EGFR) signaling in A431 cells through S-oxidation of its negative regulators such as protein tyrosine phosphatase (PTP) 1B. 9,10-phenanthrenequinone 35-42 epidermal growth factor receptor Homo sapiens 58-90 32493877-3 2020 The current study examined whether 9,10-PQ could activate epidermal growth factor receptor (EGFR) signaling in A431 cells through S-oxidation of its negative regulators such as protein tyrosine phosphatase (PTP) 1B. 9,10-phenanthrenequinone 35-42 epidermal growth factor receptor Homo sapiens 92-96 32493877-4 2020 9,10-PQ oxidized recombinant human PTP1B at Cys215 and inhibited its catalytic activity, an effect that was blocked by catalase (CAT), whereas cis-9,10-dihydroxy-9,10-dihydrophenanthrene (DDP), which lacks redox cycling activity, had no effect on PTP1B activity. 9,10-phenanthrenequinone 0-7 protein tyrosine phosphatase non-receptor type 1 Homo sapiens 35-40 32493877-4 2020 9,10-PQ oxidized recombinant human PTP1B at Cys215 and inhibited its catalytic activity, an effect that was blocked by catalase (CAT), whereas cis-9,10-dihydroxy-9,10-dihydrophenanthrene (DDP), which lacks redox cycling activity, had no effect on PTP1B activity. 9,10-phenanthrenequinone 0-7 catalase Homo sapiens 119-127 32493877-4 2020 9,10-PQ oxidized recombinant human PTP1B at Cys215 and inhibited its catalytic activity, an effect that was blocked by catalase (CAT), whereas cis-9,10-dihydroxy-9,10-dihydrophenanthrene (DDP), which lacks redox cycling activity, had no effect on PTP1B activity. 9,10-phenanthrenequinone 0-7 catalase Homo sapiens 129-132 32493877-4 2020 9,10-PQ oxidized recombinant human PTP1B at Cys215 and inhibited its catalytic activity, an effect that was blocked by catalase (CAT), whereas cis-9,10-dihydroxy-9,10-dihydrophenanthrene (DDP), which lacks redox cycling activity, had no effect on PTP1B activity. 9,10-phenanthrenequinone 0-7 protein tyrosine phosphatase non-receptor type 1 Homo sapiens 247-252 32493877-5 2020 Exposure of A431 cells to 9,10-PQ, but not DDP, activated signaling through EGFR and its downstream extracellular signal-regulated kinase 1/2 (ERK1/2), coupled with a decrease of cellular PTP activity. 9,10-phenanthrenequinone 26-33 epidermal growth factor receptor Homo sapiens 76-80 32493877-5 2020 Exposure of A431 cells to 9,10-PQ, but not DDP, activated signaling through EGFR and its downstream extracellular signal-regulated kinase 1/2 (ERK1/2), coupled with a decrease of cellular PTP activity. 9,10-phenanthrenequinone 26-33 mitogen-activated protein kinase 1 Homo sapiens 100-141 32493877-5 2020 Exposure of A431 cells to 9,10-PQ, but not DDP, activated signaling through EGFR and its downstream extracellular signal-regulated kinase 1/2 (ERK1/2), coupled with a decrease of cellular PTP activity. 9,10-phenanthrenequinone 26-33 mitogen-activated protein kinase 3 Homo sapiens 143-149 32493877-6 2020 Immunoprecipitation and UPLC-MSE revealed that PTP1B easily undergoes oxidation during exposure of A431 cells to 9,10-PQ. 9,10-phenanthrenequinone 113-120 protein tyrosine phosphatase non-receptor type 1 Homo sapiens 47-52 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 78-85 catalase Homo sapiens 54-57 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 78-85 catalase Homo sapiens 59-66 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 181-188 catalase Homo sapiens 54-57 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 181-188 catalase Homo sapiens 59-66 32493877-7 2020 Pretreatment with polyethylene glycol conjugated with CAT (PEG-CAT) abolished 9,10-PQ-generated H2O2 production and significantly blocked the activation of EGFR-ERK1/2 signaling by 9,10-PQ, indicating the involvement of H2O2 in the activation because scavenging agents for hydroxyl radicals had no effect on the redox signal activation. 9,10-phenanthrenequinone 181-188 mitogen-activated protein kinase 3 Homo sapiens 161-167 33268681-0 2020 Redox cycling of 9,10-phenanthrenequinone activates epidermal growth factor receptor signaling through S-oxidation of protein tyrosine phosphatase 1B. 9,10-phenanthrenequinone 17-41 epidermal growth factor receptor Homo sapiens 52-84 29108775-0 2018 Facilitation of 9,10-phenanthrenequinone-elicited neuroblastoma cell apoptosis by NAD(P)H:quinone oxidoreductase 1. 9,10-phenanthrenequinone 16-40 NAD(P)H quinone dehydrogenase 1 Homo sapiens 82-114 31357365-7 2019 For quinones, it varied from 1.12 mug L-1 (1,4-naphthoquinone) to 1.70 mug L-1(9,10-phenanthrenequinone). 9,10-phenanthrenequinone 79-103 immunoglobulin kappa variable 1-16 Homo sapiens 75-78 30719914-4 2019 No reaction occurred when 9,10-PQ was incubated with Na2S; however, when 5 muM 9,10-PQ was incubated with either 250 muM Na2S2 or Na2S4, we detected extensive consumption of dissolved oxygen (84 muM). 9,10-phenanthrenequinone 79-86 latexin Homo sapiens 75-78 30719914-4 2019 No reaction occurred when 9,10-PQ was incubated with Na2S; however, when 5 muM 9,10-PQ was incubated with either 250 muM Na2S2 or Na2S4, we detected extensive consumption of dissolved oxygen (84 muM). 9,10-phenanthrenequinone 79-86 latexin Homo sapiens 117-120 30719914-4 2019 No reaction occurred when 9,10-PQ was incubated with Na2S; however, when 5 muM 9,10-PQ was incubated with either 250 muM Na2S2 or Na2S4, we detected extensive consumption of dissolved oxygen (84 muM). 9,10-phenanthrenequinone 79-86 latexin Homo sapiens 117-120 29108775-3 2018 SK-N-SH cell treatment with a lethal concentration of PQ facilitated ROS production within 6 h. The treatment also promoted formation of 8-hydroxy-deoxyguanosine, p53 activation, elevation of Bax/Bcl-2 ratio, lowering of mitochondrial membrane potential, and resultant activation of caspase-9 and caspase-3, inferring that ROS production, DNA damage and mitochondrial dysfunction are crucial processes of the PQ-triggered SK-N-SH cell apoptosis. 9,10-phenanthrenequinone 54-56 tumor protein p53 Homo sapiens 163-166 29108775-10 2018 Taken together, PQ exposure produces large amounts of ROS in neuroblastoma cells via NQO1 up-regulation and resultant acceleration of its redox-cycling, followed by activation of the ROS-dependent apoptotic mechanism. 9,10-phenanthrenequinone 16-18 NAD(P)H quinone dehydrogenase 1 Homo sapiens 85-89 29108775-3 2018 SK-N-SH cell treatment with a lethal concentration of PQ facilitated ROS production within 6 h. The treatment also promoted formation of 8-hydroxy-deoxyguanosine, p53 activation, elevation of Bax/Bcl-2 ratio, lowering of mitochondrial membrane potential, and resultant activation of caspase-9 and caspase-3, inferring that ROS production, DNA damage and mitochondrial dysfunction are crucial processes of the PQ-triggered SK-N-SH cell apoptosis. 9,10-phenanthrenequinone 54-56 BCL2 associated X, apoptosis regulator Homo sapiens 192-195 29108775-3 2018 SK-N-SH cell treatment with a lethal concentration of PQ facilitated ROS production within 6 h. The treatment also promoted formation of 8-hydroxy-deoxyguanosine, p53 activation, elevation of Bax/Bcl-2 ratio, lowering of mitochondrial membrane potential, and resultant activation of caspase-9 and caspase-3, inferring that ROS production, DNA damage and mitochondrial dysfunction are crucial processes of the PQ-triggered SK-N-SH cell apoptosis. 9,10-phenanthrenequinone 54-56 BCL2 apoptosis regulator Homo sapiens 196-201 29108775-3 2018 SK-N-SH cell treatment with a lethal concentration of PQ facilitated ROS production within 6 h. The treatment also promoted formation of 8-hydroxy-deoxyguanosine, p53 activation, elevation of Bax/Bcl-2 ratio, lowering of mitochondrial membrane potential, and resultant activation of caspase-9 and caspase-3, inferring that ROS production, DNA damage and mitochondrial dysfunction are crucial processes of the PQ-triggered SK-N-SH cell apoptosis. 9,10-phenanthrenequinone 54-56 caspase 9 Homo sapiens 283-292 29108775-3 2018 SK-N-SH cell treatment with a lethal concentration of PQ facilitated ROS production within 6 h. The treatment also promoted formation of 8-hydroxy-deoxyguanosine, p53 activation, elevation of Bax/Bcl-2 ratio, lowering of mitochondrial membrane potential, and resultant activation of caspase-9 and caspase-3, inferring that ROS production, DNA damage and mitochondrial dysfunction are crucial processes of the PQ-triggered SK-N-SH cell apoptosis. 9,10-phenanthrenequinone 54-56 caspase 3 Homo sapiens 297-306 28992312-6 2017 Linoleic and oleic acids also inhibited AKR1C3-mediated metabolism of 9,10-phenanthrenequinone in colon DLD1 cells. 9,10-phenanthrenequinone 70-94 aldo-keto reductase family 1 member C3 Homo sapiens 40-46 32264381-5 2017 On the other hand, modification of an electron-withdrawing moiety such as an indolium moiety on the same phenanthrenequinone imidazole-core provided a new material PID, which exhibited favorable TP emission, indicating that phenanthrenequinone imidazole derivatives could be exploited as TP materials. 9,10-phenanthrenequinone 105-124 metastasis associated 1 family member 2 Homo sapiens 164-167 32264381-5 2017 On the other hand, modification of an electron-withdrawing moiety such as an indolium moiety on the same phenanthrenequinone imidazole-core provided a new material PID, which exhibited favorable TP emission, indicating that phenanthrenequinone imidazole derivatives could be exploited as TP materials. 9,10-phenanthrenequinone 224-243 metastasis associated 1 family member 2 Homo sapiens 164-167 28805980-0 2017 An environmental pollutant, 9,10-phenanthrenequinone, activates human TRPA1 via critical cysteines 621 and 665. 9,10-phenanthrenequinone 28-52 transient receptor potential cation channel subfamily A member 1 Homo sapiens 70-75 28805980-2 2017 In our recent study, 9, 10-phenanthrenequinone (9,10-PQ) is the most potent irritant for activation of NRF2 among 1395 cigarette smoke components and it may be, therefore, important to find its additional targets. 9,10-phenanthrenequinone 21-46 NFE2 like bZIP transcription factor 2 Homo sapiens 103-107 28805980-2 2017 In our recent study, 9, 10-phenanthrenequinone (9,10-PQ) is the most potent irritant for activation of NRF2 among 1395 cigarette smoke components and it may be, therefore, important to find its additional targets. 9,10-phenanthrenequinone 48-55 NFE2 like bZIP transcription factor 2 Homo sapiens 103-107 28805980-3 2017 Here, we show that 9,10-PQ functions as an activator of TRPA1 in human embryonic kidney (HEK) cells expressing human wild-type TRPA1 (HEK-wTRPA1) and human alveolar A549 (A549) cells. 9,10-phenanthrenequinone 19-26 transient receptor potential cation channel subfamily A member 1 Homo sapiens 56-61 28805980-3 2017 Here, we show that 9,10-PQ functions as an activator of TRPA1 in human embryonic kidney (HEK) cells expressing human wild-type TRPA1 (HEK-wTRPA1) and human alveolar A549 (A549) cells. 9,10-phenanthrenequinone 19-26 transient receptor potential cation channel subfamily A member 1 Homo sapiens 127-132 28805980-12 2017 Our findings demonstrate that 9,10-PQ activation of human TRA1 is dependent on cysteine residues 621 and 665. 9,10-phenanthrenequinone 30-37 phospholipid scramblase 4 Homo sapiens 58-62 28595002-6 2017 Using 9,10-phenanthrenequinone as the substrate, quinone redox cycling was found to inhibit DCXR reduction of l-xylulose and diacetyl. 9,10-phenanthrenequinone 6-30 dicarbonyl and L-xylulose reductase Homo sapiens 92-96 27103119-3 2016 In comparison, the reaction of trans-[Ru(III)(PQ (-))(PPh3)2Cl2] (PQ), a 9,10-phenanthrenequinone (PQ) analogue of affords only trans-[Ru(III)(PQ)(PPh3)2Cl2](+)Br3(-) ((+)Br3(-)). 9,10-phenanthrenequinone 46-48 caveolin 1 Homo sapiens 54-58 29187913-8 2017 The o-MOCA was optimized and its performance evaluated using the 9,10-phenanthraquinone (PQ) as a standard redox-active compound. 9,10-phenanthrenequinone 65-87 dedicator of cytokinesis 3 Homo sapiens 6-10 29187913-8 2017 The o-MOCA was optimized and its performance evaluated using the 9,10-phenanthraquinone (PQ) as a standard redox-active compound. 9,10-phenanthrenequinone 89-91 dedicator of cytokinesis 3 Homo sapiens 6-10 29187913-9 2017 Laboratory testing shows minimum interferences or carryover between consecutive samples, low blanks, and a reproducible, linear response between the DTT consumption rate (nmol min-1) and PQ concentration (muM). 9,10-phenanthrenequinone 187-189 CD59 molecule (CD59 blood group) Homo sapiens 176-181 29187913-9 2017 Laboratory testing shows minimum interferences or carryover between consecutive samples, low blanks, and a reproducible, linear response between the DTT consumption rate (nmol min-1) and PQ concentration (muM). 9,10-phenanthrenequinone 187-189 latexin Homo sapiens 205-208 27103119-3 2016 In comparison, the reaction of trans-[Ru(III)(PQ (-))(PPh3)2Cl2] (PQ), a 9,10-phenanthrenequinone (PQ) analogue of affords only trans-[Ru(III)(PQ)(PPh3)2Cl2](+)Br3(-) ((+)Br3(-)). 9,10-phenanthrenequinone 46-48 caveolin 1 Homo sapiens 147-151 27103119-3 2016 In comparison, the reaction of trans-[Ru(III)(PQ (-))(PPh3)2Cl2] (PQ), a 9,10-phenanthrenequinone (PQ) analogue of affords only trans-[Ru(III)(PQ)(PPh3)2Cl2](+)Br3(-) ((+)Br3(-)). 9,10-phenanthrenequinone 73-97 caveolin 1 Homo sapiens 54-58 27103119-3 2016 In comparison, the reaction of trans-[Ru(III)(PQ (-))(PPh3)2Cl2] (PQ), a 9,10-phenanthrenequinone (PQ) analogue of affords only trans-[Ru(III)(PQ)(PPh3)2Cl2](+)Br3(-) ((+)Br3(-)). 9,10-phenanthrenequinone 66-68 caveolin 1 Homo sapiens 54-58 27103119-3 2016 In comparison, the reaction of trans-[Ru(III)(PQ (-))(PPh3)2Cl2] (PQ), a 9,10-phenanthrenequinone (PQ) analogue of affords only trans-[Ru(III)(PQ)(PPh3)2Cl2](+)Br3(-) ((+)Br3(-)). 9,10-phenanthrenequinone 66-68 caveolin 1 Homo sapiens 147-151 24273736-3 2013 In this study, we examined feasibility of using tanshinones, a novel class of phenanthrenequinone-based cytoprotective Nrf2 inducers derived from the medicinal plant Salvia miltiorrhiza, for protection of cultured human skin cells and reconstructed human skin against solar simulated UV. 9,10-phenanthrenequinone 78-97 NFE2 like bZIP transcription factor 2 Homo sapiens 119-123 24646012-9 2014 The identification of 9,10-catechol conjugates supports metabolic detoxification of phenanthrene-9,10-quinone through interception of redox cycling by UGT, COMT, and SULT isozymes and indicates the possible use of phenanthrene-9,10-catechol conjugates as biomarkers of human exposure to oxygenated PAH. 9,10-phenanthrenequinone 84-109 UDP glucuronosyltransferase family 1 member A complex locus Homo sapiens 151-154 24646012-9 2014 The identification of 9,10-catechol conjugates supports metabolic detoxification of phenanthrene-9,10-quinone through interception of redox cycling by UGT, COMT, and SULT isozymes and indicates the possible use of phenanthrene-9,10-catechol conjugates as biomarkers of human exposure to oxygenated PAH. 9,10-phenanthrenequinone 84-109 catechol-O-methyltransferase Homo sapiens 156-160 26381591-3 2015 Therefore, a comprehensive analysis of the conformational structure and the solvent polarity induced energy level reordering of the two lowest triplet states of 9,10-phenanthrenequinone (PQ) was carried out using nanosecond-time-resolved absorption (ns-TRA), time-resolved resonance Raman (TR(3)) spectroscopy, and time dependent-density functional theory (TD-DFT) studies. 9,10-phenanthrenequinone 161-185 T cell receptor alpha locus Homo sapiens 253-256 26381591-3 2015 Therefore, a comprehensive analysis of the conformational structure and the solvent polarity induced energy level reordering of the two lowest triplet states of 9,10-phenanthrenequinone (PQ) was carried out using nanosecond-time-resolved absorption (ns-TRA), time-resolved resonance Raman (TR(3)) spectroscopy, and time dependent-density functional theory (TD-DFT) studies. 9,10-phenanthrenequinone 187-189 T cell receptor alpha locus Homo sapiens 253-256 25550200-2 2015 SPR also mediates chemical redox cycling, catalyzing one-electron reduction of redox-active chemicals, including quinones and bipyridinium herbicides (e.g., menadione, 9,10-phenanthrenequinone, and diquat); rapid reaction of the reduced radicals with molecular oxygen generates reactive oxygen species (ROS). 9,10-phenanthrenequinone 168-192 sepiapterin reductase Rattus norvegicus 0-3 26190959-7 2014 A comprehensive analysis of the coupled chemo-enzymatic reactions revealed pronounced inhibition of hUGDH by NADH and UDP-xylose as well as an adequate oxygen supply for PQ re-oxidation as major bottlenecks of effective performance of the overall multi-step reaction system. 9,10-phenanthrenequinone 170-172 UDP-glucose 6-dehydrogenase Homo sapiens 100-105 23640889-4 2013 In addition to reducing sepiapterin, SPR mediated chemical redox cycling of bipyridinium herbicides and various quinones; this activity was greatest for 1,2-naphthoquinone followed by 9,10-phenanthrenequinone, 1,4-naphthoquinone, menadione, and 2,3-dimethyl-1,4-naphthoquinone. 9,10-phenanthrenequinone 184-208 sepiapterin reductase Homo sapiens 37-40 23475264-1 2013 Reactions of 9,10-phenanthrenequinone (PQ) in toluene with [M(II)(PPh3)3X2] at 298 K afford green complexes, trans-[M(PQ)(PPh3)2X2] (M = Ru, X = Cl, 1; M = Os, X = Br, 2) in moderate yields. 9,10-phenanthrenequinone 13-37 caveolin 1 Homo sapiens 66-70 23475264-1 2013 Reactions of 9,10-phenanthrenequinone (PQ) in toluene with [M(II)(PPh3)3X2] at 298 K afford green complexes, trans-[M(PQ)(PPh3)2X2] (M = Ru, X = Cl, 1; M = Os, X = Br, 2) in moderate yields. 9,10-phenanthrenequinone 13-37 caveolin 1 Homo sapiens 122-126 23475264-1 2013 Reactions of 9,10-phenanthrenequinone (PQ) in toluene with [M(II)(PPh3)3X2] at 298 K afford green complexes, trans-[M(PQ)(PPh3)2X2] (M = Ru, X = Cl, 1; M = Os, X = Br, 2) in moderate yields. 9,10-phenanthrenequinone 39-41 caveolin 1 Homo sapiens 66-70 23475264-1 2013 Reactions of 9,10-phenanthrenequinone (PQ) in toluene with [M(II)(PPh3)3X2] at 298 K afford green complexes, trans-[M(PQ)(PPh3)2X2] (M = Ru, X = Cl, 1; M = Os, X = Br, 2) in moderate yields. 9,10-phenanthrenequinone 39-41 caveolin 1 Homo sapiens 122-126 23475264-2 2013 Reaction of anhydrous RhCl3 with PQ and PPh3 in boiling ethanol affords the dark brown paramagnetic complex, cis-[Rh(PQ)(PPh3)2Cl2] (3) in good yields. 9,10-phenanthrenequinone 33-35 caveolin 1 Homo sapiens 121-125 22925602-2 2012 9,10-Phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust particles, produces ROS in redox cycling following two-electron reduction by NAD(P)H:quinone oxidoreductase 1 (NQO1), which has been considered as a cause of its cyto- and genotoxicity. 9,10-phenanthrenequinone 0-24 NAD(P)H quinone dehydrogenase 1 Homo sapiens 147-179 22925602-2 2012 9,10-Phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust particles, produces ROS in redox cycling following two-electron reduction by NAD(P)H:quinone oxidoreductase 1 (NQO1), which has been considered as a cause of its cyto- and genotoxicity. 9,10-phenanthrenequinone 0-24 NAD(P)H quinone dehydrogenase 1 Homo sapiens 181-185 22925602-2 2012 9,10-Phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust particles, produces ROS in redox cycling following two-electron reduction by NAD(P)H:quinone oxidoreductase 1 (NQO1), which has been considered as a cause of its cyto- and genotoxicity. 9,10-phenanthrenequinone 26-33 NAD(P)H quinone dehydrogenase 1 Homo sapiens 147-179 22925602-2 2012 9,10-Phenanthrenequinone (9,10-PQ), a major quinone in diesel exhaust particles, produces ROS in redox cycling following two-electron reduction by NAD(P)H:quinone oxidoreductase 1 (NQO1), which has been considered as a cause of its cyto- and genotoxicity. 9,10-phenanthrenequinone 26-33 NAD(P)H quinone dehydrogenase 1 Homo sapiens 181-185 22925602-3 2012 In this study, we show that NAC unexpectedly augments the toxicity of 9,10-PQ in cells with low NQO1 activity. 9,10-phenanthrenequinone 70-77 X-linked Kx blood group Homo sapiens 28-31 22925602-3 2012 In this study, we show that NAC unexpectedly augments the toxicity of 9,10-PQ in cells with low NQO1 activity. 9,10-phenanthrenequinone 70-77 NAD(P)H quinone dehydrogenase 1 Homo sapiens 96-100 22925602-5 2012 In the skin cells, the cytotoxicity of 9,10-PQ was significantly enhanced by addition of NAC. 9,10-phenanthrenequinone 39-46 X-linked Kx blood group Homo sapiens 89-92 22925602-9 2012 The results suggested that dual effects of NAC on the cyto- and genotoxicity of 9,10-PQ were dependent on tissue-specific NQO1 activity. 9,10-phenanthrenequinone 80-87 X-linked Kx blood group Homo sapiens 43-46 22925602-9 2012 The results suggested that dual effects of NAC on the cyto- and genotoxicity of 9,10-PQ were dependent on tissue-specific NQO1 activity. 9,10-phenanthrenequinone 80-87 NAD(P)H quinone dehydrogenase 1 Homo sapiens 122-126 22281686-8 2012 The exposure of A549 cells to 9,10-PQ provokes viability loss, which is significantly protected by the addition of the AKR1C3 inhibitor and antioxidant enzyme and by the removal of the surfactants from the culture medium. 9,10-phenanthrenequinone 30-37 aldo-keto reductase family 1 member C3 Homo sapiens 119-125 22975515-4 2012 Flow cytometric analyses of U937 cells after long-term exposure to 9,10-PQ revealed induction of differentiation that was evidenced by increasing expression of CD11b/CD18, a cell-surface marker for monocytic differentiation into macrophages. 9,10-phenanthrenequinone 67-74 integrin subunit alpha M Homo sapiens 160-165 22975515-4 2012 Flow cytometric analyses of U937 cells after long-term exposure to 9,10-PQ revealed induction of differentiation that was evidenced by increasing expression of CD11b/CD18, a cell-surface marker for monocytic differentiation into macrophages. 9,10-phenanthrenequinone 67-74 integrin subunit beta 2 Homo sapiens 166-170 22975515-6 2012 The 9,10-PQ treatment increased the expression of aldo-keto reductase (AKR) 1C3, which reached a peak at 1 to 2 d post-treatment and then declined. 9,10-phenanthrenequinone 4-11 aldo-keto reductase family 1 member C3 Homo sapiens 50-79 22975515-7 2012 The bell-shaped curve of the AKR1C3 expression by 9,10-PQ resembled that caused by phorbol 12-myristate 13-acetate, a differentiation inducer. 9,10-phenanthrenequinone 50-57 aldo-keto reductase family 1 member C3 Homo sapiens 29-35 22975515-8 2012 Additionally, the concomitant treatment with tolfenamic acid, a selective AKR1C3 inhibitor, sensitized the differentiation induced by 9,10-PQ. 9,10-phenanthrenequinone 134-141 aldo-keto reductase family 1 member C3 Homo sapiens 74-80 22975515-9 2012 These results suggest that ROS formation during 9,10-PQ treatment acutely leads to apoptosis of U937 cells and the initiation of monocytic differentiation, which proceeds after the provisional overexpression of AKR1C3. 9,10-phenanthrenequinone 48-55 aldo-keto reductase family 1 member C3 Homo sapiens 211-217 18571493-4 2008 Human DHRS4 reduced various aromatic ketones and alpha-dicarbonyl compounds including cytotoxic 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 96-120 dehydrogenase/reductase 4 Homo sapiens 6-11 21623689-3 2011 The 9,10-PQ-mediated CHOP induction was strengthened by a proteasome inhibitor (MG132) and the MG132-induced cell sensitization to the 9,10-PQ toxicity was abolished by a ROS inhibitor, suggesting that ROS generation and consequent proteasomal dysfunction are responsible for the CHOP up-regulation caused by 9,10-PQ. 9,10-phenanthrenequinone 4-11 DNA-damage inducible transcript 3 Rattus norvegicus 21-25 21623689-3 2011 The 9,10-PQ-mediated CHOP induction was strengthened by a proteasome inhibitor (MG132) and the MG132-induced cell sensitization to the 9,10-PQ toxicity was abolished by a ROS inhibitor, suggesting that ROS generation and consequent proteasomal dysfunction are responsible for the CHOP up-regulation caused by 9,10-PQ. 9,10-phenanthrenequinone 4-11 DNA-damage inducible transcript 3 Rattus norvegicus 280-284 21623689-3 2011 The 9,10-PQ-mediated CHOP induction was strengthened by a proteasome inhibitor (MG132) and the MG132-induced cell sensitization to the 9,10-PQ toxicity was abolished by a ROS inhibitor, suggesting that ROS generation and consequent proteasomal dysfunction are responsible for the CHOP up-regulation caused by 9,10-PQ. 9,10-phenanthrenequinone 135-142 DNA-damage inducible transcript 3 Rattus norvegicus 280-284 21623689-3 2011 The 9,10-PQ-mediated CHOP induction was strengthened by a proteasome inhibitor (MG132) and the MG132-induced cell sensitization to the 9,10-PQ toxicity was abolished by a ROS inhibitor, suggesting that ROS generation and consequent proteasomal dysfunction are responsible for the CHOP up-regulation caused by 9,10-PQ. 9,10-phenanthrenequinone 135-142 DNA-damage inducible transcript 3 Rattus norvegicus 280-284 19442656-0 2009 Involvement of an aldo-keto reductase (AKR1C3) in redox cycling of 9,10-phenanthrenequinone leading to apoptosis in human endothelial cells. 9,10-phenanthrenequinone 67-91 aldo-keto reductase family 1 member C3 Homo sapiens 39-45 19442656-4 2009 Comparison of mRNA expression levels and kinetic constants in the 9,10-PQ reduction among 10 human reductases suggests that aldo-keto reductase 1C3 (AKR1C3) is a 9,10-PQ reductase in HAECs. 9,10-phenanthrenequinone 66-73 aldo-keto reductase family 1 member C3 Homo sapiens 124-147 19442656-4 2009 Comparison of mRNA expression levels and kinetic constants in the 9,10-PQ reduction among 10 human reductases suggests that aldo-keto reductase 1C3 (AKR1C3) is a 9,10-PQ reductase in HAECs. 9,10-phenanthrenequinone 66-73 aldo-keto reductase family 1 member C3 Homo sapiens 149-155 19442656-5 2009 In in vitro 9,10-PQ reduction by AKR1C3, the reduced product 9,10-dihydroxyphenanthrene and superoxide anions were formed, suggesting the enzymatic two-electron reduction of 9,10-PQ that thereby causes oxidative stress through its redox cycling. 9,10-phenanthrenequinone 12-19 aldo-keto reductase family 1 member C3 Homo sapiens 33-39 19442656-5 2009 In in vitro 9,10-PQ reduction by AKR1C3, the reduced product 9,10-dihydroxyphenanthrene and superoxide anions were formed, suggesting the enzymatic two-electron reduction of 9,10-PQ that thereby causes oxidative stress through its redox cycling. 9,10-phenanthrenequinone 174-181 aldo-keto reductase family 1 member C3 Homo sapiens 33-39 19442656-6 2009 In addition, the participation of AKR1C3 in 9,10-PQ-redox cycling was confirmed by the data that AKR1C3 overexpression in endothelial cells augmented the ROS generation and cytotoxicity by 9,10-PQ, and the ROS scavengers inhibited the toxic effects. 9,10-phenanthrenequinone 44-51 aldo-keto reductase family 1 member C3 Homo sapiens 34-40 19442656-6 2009 In addition, the participation of AKR1C3 in 9,10-PQ-redox cycling was confirmed by the data that AKR1C3 overexpression in endothelial cells augmented the ROS generation and cytotoxicity by 9,10-PQ, and the ROS scavengers inhibited the toxic effects. 9,10-phenanthrenequinone 44-51 aldo-keto reductase family 1 member C3 Homo sapiens 97-103 19442656-6 2009 In addition, the participation of AKR1C3 in 9,10-PQ-redox cycling was confirmed by the data that AKR1C3 overexpression in endothelial cells augmented the ROS generation and cytotoxicity by 9,10-PQ, and the ROS scavengers inhibited the toxic effects. 9,10-phenanthrenequinone 189-196 aldo-keto reductase family 1 member C3 Homo sapiens 34-40 19442656-6 2009 In addition, the participation of AKR1C3 in 9,10-PQ-redox cycling was confirmed by the data that AKR1C3 overexpression in endothelial cells augmented the ROS generation and cytotoxicity by 9,10-PQ, and the ROS scavengers inhibited the toxic effects. 9,10-phenanthrenequinone 189-196 aldo-keto reductase family 1 member C3 Homo sapiens 97-103 19442656-7 2009 Pretreatment of the overexpressing cells with AKR1C3 inhibitors, flufenamic acid and indomethacin, suppressed the 9,10-PQ-induced GSH depletion. 9,10-phenanthrenequinone 114-121 aldo-keto reductase family 1 member C3 Homo sapiens 46-52 19442656-8 2009 These results suggest that AKR1C3 is a key enzyme in the initial step of 9,10-PQ-induced cytotoxicity in HAECs. 9,10-phenanthrenequinone 73-80 aldo-keto reductase family 1 member C3 Homo sapiens 27-33 19000905-6 2008 We further demonstrate that the in vitro quinone reduction by CBR4 generates superoxide through the redox cycling, and suggest that the enzyme may be involved in the induction of apoptosis by cytotoxic 9,10-phenanthrenequinone. 9,10-phenanthrenequinone 202-226 carbonyl reductase 4 Homo sapiens 62-66 18294972-10 2008 In addition, deletion of the transcription factor Nrf2 decreased PQHG formation and increased PQ-mediated toxicity of mouse primary hepatocytes. 9,10-phenanthrenequinone 65-67 nuclear factor, erythroid derived 2, like 2 Mus musculus 50-54 18206670-0 2008 L-Xylulose reductase is involved in 9,10-phenanthrenequinone-induced apoptosis in human T lymphoma cells. 9,10-phenanthrenequinone 36-60 dicarbonyl and L-xylulose reductase Homo sapiens 0-20 18206670-6 2008 Surprisingly, the ROS generation and cytotoxicity by 9,10-PQ were augmented in an XR-transformed cell line. 9,10-phenanthrenequinone 53-60 dicarbonyl and L-xylulose reductase Homo sapiens 82-84 18206670-7 2008 XR indeed reduced 9,10-PQ and produced superoxide anion through redox cycling. 9,10-phenanthrenequinone 18-25 dicarbonyl and L-xylulose reductase Homo sapiens 0-2 18206670-9 2008 Moreover, the 9,10-PQ-induced apoptosis was partially inhibited by the pretreatment with XR-specific inhibitors. 9,10-phenanthrenequinone 14-21 dicarbonyl and L-xylulose reductase Homo sapiens 89-91 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 72-96 aldo-keto reductase family 1 member C3 Homo sapiens 115-121 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 72-96 aldo-keto reductase family 1 member C1 Homo sapiens 155-161 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 72-96 aldo-keto reductase family 1 member C2 Homo sapiens 166-172 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 98-100 aldo-keto reductase family 1 member C3 Homo sapiens 115-121 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 98-100 aldo-keto reductase family 1 member C1 Homo sapiens 155-161 17950253-4 2008 We tested NSAIDs and NSAID analogues for inhibition of the reduction of 9,10-phenanthrenequinone (PQ) catalyzed by AKR1C3 and the closely related isoforms AKR1C1 and AKR1C2. 9,10-phenanthrenequinone 98-100 aldo-keto reductase family 1 member C2 Homo sapiens 166-172 17950253-9 2008 The pattern of inhibition of AKR1C3 by indomethacin and CBM was uncompetitive versus PQ, but competitive versus Delta(4)-androstene-3,17-dione, indicating that two different inhibitory complexes form during the ordered bi bi reactions. 9,10-phenanthrenequinone 85-87 aldo-keto reductase family 1 member C3 Homo sapiens 29-35