PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 28905270-3 2018 The findings were that diabetes induced by both doses of alloxan was accompanied by significant increases in the lysosomal activities of acid phosphatase and the glycosidases investigated: beta-glucuronidase, beta-galactosidase, beta-glucosidase, and N-acetyl-hexosaminidase. Alloxan 57-64 glucuronidase, beta Mus musculus 189-207 28816409-7 2018 In vivo, genistein also reduced fasting glucose levels accompanied with reduced PEPCK-C expression and increased in AMPK and ERK1/2 phosphorylation states in the liver of genistein-treated alloxan-induced diabetic mice. Alloxan 189-196 protein kinase AMP-activated catalytic subunit alpha 1 Homo sapiens 116-120 28905270-3 2018 The findings were that diabetes induced by both doses of alloxan was accompanied by significant increases in the lysosomal activities of acid phosphatase and the glycosidases investigated: beta-glucuronidase, beta-galactosidase, beta-glucosidase, and N-acetyl-hexosaminidase. Alloxan 57-64 galactosidase, beta 1 Mus musculus 209-227 28842434-5 2017 In response to an acute bolus intravenous injection of ANG II, alloxan-induced diabetic mice exhibited a higher mean arterial pressure (MAP) (119.1 +- 3.8 vs. 106.2 +- 3.5 mmHg) and a lower renal blood flow (0.25 +- 0.07 vs. 0.52 +- 0.14 ml/min) compared with nondiabetic mice. Alloxan 63-70 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 55-61 28837876-7 2017 Importantly, phthalhydrazide treatment of alloxan-treated diabetic rats markedly decreased the concentrationof interleukin-6 (IL-6) and corticosterone level. Alloxan 42-49 interleukin 6 Rattus norvegicus 111-124 28837876-7 2017 Importantly, phthalhydrazide treatment of alloxan-treated diabetic rats markedly decreased the concentrationof interleukin-6 (IL-6) and corticosterone level. Alloxan 42-49 interleukin 6 Rattus norvegicus 126-130 28423281-9 2017 Alloxan induced degenerative changes in hepatic and pancreatic tissues, increased hepatic lipid peroxidation, and increased gene expression of PC and caspase 3. Alloxan 0-7 caspase 3 Rattus norvegicus 150-159 28802580-7 2017 The repressive activity of BAP1 was relieved by alloxan but exacerbated by PUGNAc, an O-GlcNAcase (OGA) inhibitor. Alloxan 48-55 Brca1 associated protein 1 Mus musculus 27-31 27114244-0 2016 Corrigendum to "BDNF protects pancreatic beta cells (RIN5F) against cytotoxic action of alloxan, streptozotocin, doxorubicin and benzo(a)pyrene in vitro" METABOLISM CLINICAL AND EXPERIMENTAL 65 (2016) 667-684. Alloxan 88-95 brain derived neurotrophic factor Homo sapiens 16-20 28190920-6 2017 Eight hours after AL treatment, aquaporin 1-negative and Na/K pump-positive thick ascending limbs of Henle (TAL) were degenerated in the outer medulla. Alloxan 18-20 aquaporin 1 Rattus norvegicus 32-43 27776166-8 2016 In addition, GlcN-mediated O-GlcNAc modification was involved in the downregulation of TNF-alpha and IL-8 genes but not IL-6 and IL-24 genes, based on the effects of alloxan, an O-GlcNAc transferase inhibitor. Alloxan 166-173 tumor necrosis factor Homo sapiens 87-96 26908204-6 2016 Oral administration in the alloxan-induced diabetic mice of the mixture loaded in elastic anionic niosomes with the insulin doses at 25, 50 and 100 IU/kg body weight indicated significant hypoglycemic activity with the percentage fasting blood glucose reduction of 1.95, 2.10 and 2.10 folds of the subcutaneous insulin injection at 12 h, respectively. Alloxan 27-34 insulin Homo sapiens 116-123 26908204-6 2016 Oral administration in the alloxan-induced diabetic mice of the mixture loaded in elastic anionic niosomes with the insulin doses at 25, 50 and 100 IU/kg body weight indicated significant hypoglycemic activity with the percentage fasting blood glucose reduction of 1.95, 2.10 and 2.10 folds of the subcutaneous insulin injection at 12 h, respectively. Alloxan 27-34 insulin Homo sapiens 311-318 28220578-3 2017 Here, we hypothesize that "Alloxan-induced renal damage is associated with alterations of p53, TGF-beta1, and extracellular matrix metalloproteinases." Alloxan 27-34 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 90-93 28220578-3 2017 Here, we hypothesize that "Alloxan-induced renal damage is associated with alterations of p53, TGF-beta1, and extracellular matrix metalloproteinases." Alloxan 27-34 transforming growth factor, beta 1 Rattus norvegicus 95-104 27748816-3 2016 The current study investigated the effects of this compound (referred to as compound K16) on diabetes using an alloxan-induced diabetic mouse model. Alloxan 111-118 keratin 16 Mus musculus 85-88 26452142-10 2015 An important fraction of the beta cells significantly lost insulin positivity after alloxan administration, which was restored to normal after one week of EGF/G treatment. Alloxan 84-91 epidermal growth factor Mus musculus 155-158 27085775-1 2016 OBJECTIVE: The study was conducted to observe whether brain-derived neurotrophic factor (BDNF) has cytoprotective actions against alloxan (AL), streptozotocin (STZ), doxorubicin (DB) and benzo(a)pyrene (BP) compounds in vitro that may account for its beneficial action in diabetes mellitus. Alloxan 130-137 brain derived neurotrophic factor Mus musculus 54-87 27085775-1 2016 OBJECTIVE: The study was conducted to observe whether brain-derived neurotrophic factor (BDNF) has cytoprotective actions against alloxan (AL), streptozotocin (STZ), doxorubicin (DB) and benzo(a)pyrene (BP) compounds in vitro that may account for its beneficial action in diabetes mellitus. Alloxan 130-137 brain derived neurotrophic factor Mus musculus 89-93 27085775-1 2016 OBJECTIVE: The study was conducted to observe whether brain-derived neurotrophic factor (BDNF) has cytoprotective actions against alloxan (AL), streptozotocin (STZ), doxorubicin (DB) and benzo(a)pyrene (BP) compounds in vitro that may account for its beneficial action in diabetes mellitus. Alloxan 139-141 brain derived neurotrophic factor Mus musculus 54-87 27085775-1 2016 OBJECTIVE: The study was conducted to observe whether brain-derived neurotrophic factor (BDNF) has cytoprotective actions against alloxan (AL), streptozotocin (STZ), doxorubicin (DB) and benzo(a)pyrene (BP) compounds in vitro that may account for its beneficial action in diabetes mellitus. Alloxan 139-141 brain derived neurotrophic factor Mus musculus 89-93 27085775-5 2016 Effect of alloxan, STZ, doxorubicin and BP on the production of BDNF by RIN5F cells was also studied. Alloxan 10-17 brain derived neurotrophic factor Mus musculus 64-68 27085775-6 2016 RESULTS: Results of the present study revealed that BDNF in the doses (100ng>50ng>10ng/ml) has significant cytoprotection (P<0.001, P<0.01) on cytotoxic action of AL, STZ, DB and BP against rat insulinoma RIN5F (5x10(4) cells/100mul) cells in vitro. Alloxan 175-177 brain-derived neurotrophic factor Rattus norvegicus 52-56 26827782-0 2016 Phycocyanin ameliorates alloxan-induced diabetes mellitus in mice: Involved in insulin signaling pathway and GK expression. Alloxan 24-31 glucokinase Mus musculus 109-111 26827782-6 2016 The results suggest that phycocyanin ameliorates alloxan-induced diabetes mellitus in mice by activating insulin signaling pathway and GK expression in pancreas and liver in diabetic mice. Alloxan 49-56 glucokinase Mus musculus 135-137 26664046-0 2015 Timosaponin B-II ameliorates diabetic nephropathy via TXNIP, mTOR, and NF-kappaB signaling pathways in alloxan-induced mice. Alloxan 103-110 thioredoxin interacting protein Mus musculus 54-59 26664046-0 2015 Timosaponin B-II ameliorates diabetic nephropathy via TXNIP, mTOR, and NF-kappaB signaling pathways in alloxan-induced mice. Alloxan 103-110 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 71-80 25549789-1 2015 The effect of streptozotocin (STZ) and alloxan (ALX) on the hepatic messenger RNA (mRNA) expression of four transporters (Mrp2, Mdr1, Oct1, and Oatp1) was studied in the present work. Alloxan 39-46 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 144-149 25549789-1 2015 The effect of streptozotocin (STZ) and alloxan (ALX) on the hepatic messenger RNA (mRNA) expression of four transporters (Mrp2, Mdr1, Oct1, and Oatp1) was studied in the present work. Alloxan 48-51 ATP binding cassette subfamily C member 2 Rattus norvegicus 122-126 25549789-3 2015 The results indicated that the mRNA expression levels of the Mrp2, Mdr1, Oct1, and Oatp1 in ALX-induced diabetic rats, as well as the hepatic mRNA expression of Mdr1 and Oatp1 in STZ-induced diabetic rats, were significantly decreased as compared with the control. Alloxan 92-95 ATP binding cassette subfamily C member 2 Rattus norvegicus 61-65 25549789-3 2015 The results indicated that the mRNA expression levels of the Mrp2, Mdr1, Oct1, and Oatp1 in ALX-induced diabetic rats, as well as the hepatic mRNA expression of Mdr1 and Oatp1 in STZ-induced diabetic rats, were significantly decreased as compared with the control. Alloxan 92-95 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 67-71 25549789-3 2015 The results indicated that the mRNA expression levels of the Mrp2, Mdr1, Oct1, and Oatp1 in ALX-induced diabetic rats, as well as the hepatic mRNA expression of Mdr1 and Oatp1 in STZ-induced diabetic rats, were significantly decreased as compared with the control. Alloxan 92-95 solute carrier family 22 member 1 Rattus norvegicus 73-77 25549789-3 2015 The results indicated that the mRNA expression levels of the Mrp2, Mdr1, Oct1, and Oatp1 in ALX-induced diabetic rats, as well as the hepatic mRNA expression of Mdr1 and Oatp1 in STZ-induced diabetic rats, were significantly decreased as compared with the control. Alloxan 92-95 solute carrier organic anion transporter family, member 1a1 Rattus norvegicus 83-88 25105335-7 2015 On the other hand, we showed that alloxan administration was accompanied by an oxidative stress status assessed by an increase of malondialdehyde (MDA) and hydrogen peroxide (H2O2) levels, as well as a depletion of sulfhydril group content (-SH) and antioxidant enzyme activities as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) in testis, epididymis, and sperm. Alloxan 34-41 catalase Rattus norvegicus 311-319 25105335-7 2015 On the other hand, we showed that alloxan administration was accompanied by an oxidative stress status assessed by an increase of malondialdehyde (MDA) and hydrogen peroxide (H2O2) levels, as well as a depletion of sulfhydril group content (-SH) and antioxidant enzyme activities as superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx) in testis, epididymis, and sperm. Alloxan 34-41 catalase Rattus norvegicus 321-324 26855973-1 2015 The objective of this study is to detrmine whether alloxan-induced diabetic Lactoferrin knockout (LFKO-/-) mice are more susceptible to periodontal disease caused by Aggregatibacter actinomycetemcomitans compared to the diabetic wild-type (WT) mice. Alloxan 51-58 lactotransferrin Mus musculus 76-87 25199622-0 2015 Probucol prevents atrial remodeling by inhibiting oxidative stress and TNF-alpha/NF-kappaB/TGF-beta signal transduction pathway in alloxan-induced diabetic rabbits. Alloxan 131-138 tumor necrosis factor Oryctolagus cuniculus 71-80 25450901-0 2014 Expression of visfatin in alloxan-induced diabetic rat testis. Alloxan 26-33 nicotinamide phosphoribosyltransferase Rattus norvegicus 14-22 24939145-6 2014 Gene expression of TNF-alpha and TGF-beta1 showed gradual increase after one and two weeks of alloxan administration as compared to the normal group. Alloxan 94-101 tumor necrosis factor Rattus norvegicus 19-28 24939145-6 2014 Gene expression of TNF-alpha and TGF-beta1 showed gradual increase after one and two weeks of alloxan administration as compared to the normal group. Alloxan 94-101 transforming growth factor, beta 1 Rattus norvegicus 33-42 24814361-2 2014 We sought to investigate the effects of peroxisome proliferator-activated receptor-gamma (PPAR-gamma) activator rosiglitazone on atrial structural remodeling and atrial fibrillation (AF) promotion in alloxan-induced diabetic rabbits. Alloxan 200-207 peroxisome proliferator-activated receptor gamma Oryctolagus cuniculus 90-100 25289966-5 2014 M. oleifera leaf extract counteracted the alloxan-induced diabetic effects in rats as it normalized the elevated serum levels of glucose, triglycerides, cholesterol, and malondialdehyde, and normalized mRNA expression of the gluconeogenic enzyme pyruvate carboxylase in hepatic tissues. Alloxan 42-49 pyruvate carboxylase Rattus norvegicus 246-266 24604151-4 2014 Supplementation of vanadium to alloxan-induced diabetic rats decreased the blood glucose levels due to hyperglycemia, inhibited the AR activity, and delayed cataract progression in a dose-dependent manner. Alloxan 31-38 aldo-keto reductase family 1 member B1 Rattus norvegicus 132-134 24599112-4 2014 In this issue of Diabetologia (Brom et al DOI 10.1007/s00125-014-3166-3) it is reported that single photon emission computed tomography (SPECT) data with (111)indium-labelled glucagon-like peptide-1 (GLP-1) receptor agonist exendin-3 correlate with the morphometric analysis of beta cell mass in a rat model of alloxan-induced diabetes. Alloxan 311-318 glucagon Rattus norvegicus 175-198 24599112-4 2014 In this issue of Diabetologia (Brom et al DOI 10.1007/s00125-014-3166-3) it is reported that single photon emission computed tomography (SPECT) data with (111)indium-labelled glucagon-like peptide-1 (GLP-1) receptor agonist exendin-3 correlate with the morphometric analysis of beta cell mass in a rat model of alloxan-induced diabetes. Alloxan 311-318 glucagon-like peptide 1 receptor Rattus norvegicus 200-215 23880309-8 2013 HO-1 expression in islets was significantly upregulated in alloxan-injected Bach1-deficient mice, whereas expression of other antioxidative enzymes, e.g., catalase, superoxide dismutase, and glutathione peroxidase, was not changed by either alloxan administration or Bach1 deficiency. Alloxan 59-66 heme oxygenase 1 Mus musculus 0-4 24711743-0 2014 The Control of Hyperglycemia by Estriol and Progesterone in Alloxan induced Type I Diabetes Mellitus Mice Model through Hepatic Insulin Synthesis. Alloxan 60-67 insulin Homo sapiens 128-135 24477542-8 2014 The oxytocin (OT) and vasopressin (VP) responses to insulin+glucose were blocked by the glucokinase inhibitor alloxan (4 mM; P <= 0.002) and the PI3K inhibitor wortmannin (50 nM; OT: P = 0.03; VP: P <= 0.002). Alloxan 110-117 arginine vasopressin Rattus norvegicus 22-33 24477542-8 2014 The oxytocin (OT) and vasopressin (VP) responses to insulin+glucose were blocked by the glucokinase inhibitor alloxan (4 mM; P <= 0.002) and the PI3K inhibitor wortmannin (50 nM; OT: P = 0.03; VP: P <= 0.002). Alloxan 110-117 arginine vasopressin Rattus norvegicus 35-37 24477542-8 2014 The oxytocin (OT) and vasopressin (VP) responses to insulin+glucose were blocked by the glucokinase inhibitor alloxan (4 mM; P <= 0.002) and the PI3K inhibitor wortmannin (50 nM; OT: P = 0.03; VP: P <= 0.002). Alloxan 110-117 glucokinase Rattus norvegicus 88-99 24477542-8 2014 The oxytocin (OT) and vasopressin (VP) responses to insulin+glucose were blocked by the glucokinase inhibitor alloxan (4 mM; P <= 0.002) and the PI3K inhibitor wortmannin (50 nM; OT: P = 0.03; VP: P <= 0.002). Alloxan 110-117 arginine vasopressin Rattus norvegicus 196-198 24331702-5 2014 To complement Rab18, and promote angiogenesis eNOS was also targeted, and this novel Rab18-eNOS therapy via a dynamically controlled "fibrin-in-fibrin" delivery system, demonstrated enhanced wound closure, by increasing functional angiogenesis and reducing inflammation, in an alloxan-induced hyperglycemic preclinical ear ulcer model of compromised wound healing. Alloxan 277-284 RAB18, member RAS oncogene family Homo sapiens 85-90 23880309-8 2013 HO-1 expression in islets was significantly upregulated in alloxan-injected Bach1-deficient mice, whereas expression of other antioxidative enzymes, e.g., catalase, superoxide dismutase, and glutathione peroxidase, was not changed by either alloxan administration or Bach1 deficiency. Alloxan 59-66 BTB and CNC homology 1, basic leucine zipper transcription factor 1 Mus musculus 76-81 23539426-2 2013 This study investigated the hypoglycemic activity of orally delivered insulin - Tat mixture in alloxan-induced diabetic mice. Alloxan 95-102 insulin Homo sapiens 70-77 23970406-6 2013 In diabetic rats, gene expression of TNF-alpha and TGF-beta1 recorded a gradual increase and marked increase was observed after one and two weeks of alloxan administration, in comparison with normal rats. Alloxan 149-156 tumor necrosis factor Rattus norvegicus 37-46 23970406-6 2013 In diabetic rats, gene expression of TNF-alpha and TGF-beta1 recorded a gradual increase and marked increase was observed after one and two weeks of alloxan administration, in comparison with normal rats. Alloxan 149-156 transforming growth factor, beta 1 Rattus norvegicus 51-60 22898325-3 2012 The working hypothesis was that alloxan-induced diabetes might modify the vascular effects of ANP in isolated rabbit renal arteries and the mechanisms involved in such actions. Alloxan 32-39 natriuretic peptides A Oryctolagus cuniculus 94-97 23138840-6 2013 ALX exposure elevated the blood glucose, serum ALP and ALT levels, the production of reactive oxygen species (ROS), and disturbed the intra-cellular antioxidant machineries. Alloxan 0-3 PDZ and LIM domain 3 Rattus norvegicus 47-50 23092809-5 2013 Studies on the mechanism of apoptosis showed that ALX accelerated the markers of mitochondrial dependent apoptotic pathway (enhanced cytochrome C release in cytosol from mitochondria, altered the expression of Bax, Bcl-2, Apaf-1, caspase-9, caspase-3). Alloxan 50-53 BCL2 associated X, apoptosis regulator Rattus norvegicus 210-213 23092809-5 2013 Studies on the mechanism of apoptosis showed that ALX accelerated the markers of mitochondrial dependent apoptotic pathway (enhanced cytochrome C release in cytosol from mitochondria, altered the expression of Bax, Bcl-2, Apaf-1, caspase-9, caspase-3). Alloxan 50-53 BCL2, apoptosis regulator Rattus norvegicus 215-220 23092809-5 2013 Studies on the mechanism of apoptosis showed that ALX accelerated the markers of mitochondrial dependent apoptotic pathway (enhanced cytochrome C release in cytosol from mitochondria, altered the expression of Bax, Bcl-2, Apaf-1, caspase-9, caspase-3). Alloxan 50-53 apoptotic peptidase activating factor 1 Rattus norvegicus 222-228 23092809-5 2013 Studies on the mechanism of apoptosis showed that ALX accelerated the markers of mitochondrial dependent apoptotic pathway (enhanced cytochrome C release in cytosol from mitochondria, altered the expression of Bax, Bcl-2, Apaf-1, caspase-9, caspase-3). Alloxan 50-53 caspase 9 Rattus norvegicus 230-239 23092809-5 2013 Studies on the mechanism of apoptosis showed that ALX accelerated the markers of mitochondrial dependent apoptotic pathway (enhanced cytochrome C release in cytosol from mitochondria, altered the expression of Bax, Bcl-2, Apaf-1, caspase-9, caspase-3). Alloxan 50-53 caspase 3 Rattus norvegicus 241-250 23667864-3 2013 The severity of hyperglycemia and hypertriglyceridemia decreased, hypercholesterolemia developed, hypotocoferolemia returned to normal, and the tendency to LPO inhibition was observed in rats 10 days after alloxan administration against the background of previous one-week treatment with insulin. Alloxan 206-213 lactoperoxidase Rattus norvegicus 156-159 23138840-10 2013 In addition, ALX exposure reciprocally regulated Bcl-2 family protein expression, disturbed mitochondrial membrane potential, and subsequently released cytochrome c from mitochondria to cytosol. Alloxan 13-16 BCL2, apoptosis regulator Rattus norvegicus 49-54 22356440-9 2012 The alloxan-induced decrease in the GK activity in islets and hepatocytes was also ameliorated by exendin-4 treatment. Alloxan 4-11 glucokinase Mus musculus 36-38 22728640-6 2012 IBP was administered orally at three doses [100, 200 and 400 mg/kg body weight] for 14 days to the diabetic mice induced by alloxan. Alloxan 124-131 trafficking protein particle complex 9 Mus musculus 0-3 22659112-6 2012 Immunohistochemical studies in alloxan induced mice demonstrated a marked increase in the immunoreactivity of nuclear transcription factor (NF-kappaB). Alloxan 31-38 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 140-149 22582095-10 2012 The study arrived at the conclusion that HC generates ROS in a redox-cycling reaction with Alx that explains the decline in viability of insulin-secreting cells, leading to reduced glucokinase phosphorylating ability, diminished insulin secretory responsiveness and cell death. Alloxan 91-94 glucokinase Rattus norvegicus 181-192 22239106-3 2012 Elevated levels of blood glucose, glycosylated Hb and TNFalpha decreased levels of plasma insulin and disturbed intra-cellular antioxidant machineries were detected in ALX exposed animals. Alloxan 168-171 tumor necrosis factor Rattus norvegicus 54-62 22239106-5 2012 Studies on the mechanism of ALX-induced diabetes showed that hyperglycemia caused disruption of mitochondrial membrane potential in the spleen, released cytochrome C in the cytosol, activated caspase 3 and ultimately led to apoptotic cell death. Alloxan 28-31 caspase 3 Rattus norvegicus 192-201 22138235-10 2012 Results also suggest that taurine could protect cardiac tissue from ALX induced apoptosis via the regulation of Bcl2 family and caspase 9/3 proteins. Alloxan 68-71 BCL2, apoptosis regulator Rattus norvegicus 112-116 21981382-3 2012 Here, we used neuronal and murine models that overexpress human tau to demonstrate that mild oxidative stress generated by alloxan suppresses several phenotypes of tauopathy. Alloxan 123-130 microtubule associated protein tau Homo sapiens 64-67 21981382-4 2012 Alloxan treatment reduced HSP90 levels and promoted proteasomal degradation of tau, c-Jun N-amino terminal kinase, and histone deacetylase (HDAC) 6. Alloxan 0-7 heat shock protein 86, pseudogene 1 Mus musculus 26-31 21981382-4 2012 Alloxan treatment reduced HSP90 levels and promoted proteasomal degradation of tau, c-Jun N-amino terminal kinase, and histone deacetylase (HDAC) 6. Alloxan 0-7 microtubule associated protein tau Homo sapiens 79-82 21981382-4 2012 Alloxan treatment reduced HSP90 levels and promoted proteasomal degradation of tau, c-Jun N-amino terminal kinase, and histone deacetylase (HDAC) 6. Alloxan 0-7 jun proto-oncogene Mus musculus 84-89 21981382-4 2012 Alloxan treatment reduced HSP90 levels and promoted proteasomal degradation of tau, c-Jun N-amino terminal kinase, and histone deacetylase (HDAC) 6. Alloxan 0-7 histone deacetylase 6 Mus musculus 119-147 21981382-6 2012 Age-dependent decreases in HDAC2 and phospho-tau levels correlated with spatial memory enhancement in alloxan-injected tau mice. Alloxan 102-109 histone deacetylase 2 Mus musculus 27-32 21981382-6 2012 Age-dependent decreases in HDAC2 and phospho-tau levels correlated with spatial memory enhancement in alloxan-injected tau mice. Alloxan 102-109 microtubule associated protein tau Homo sapiens 45-48 21981382-6 2012 Age-dependent decreases in HDAC2 and phospho-tau levels correlated with spatial memory enhancement in alloxan-injected tau mice. Alloxan 102-109 microtubule associated protein tau Homo sapiens 119-122 22138235-10 2012 Results also suggest that taurine could protect cardiac tissue from ALX induced apoptosis via the regulation of Bcl2 family and caspase 9/3 proteins. Alloxan 68-71 caspase 9 Rattus norvegicus 128-137 21946410-0 2011 IGF1/insulin receptor kinase inhibition by BMS-536924 is better tolerated than alloxan-induced hypoinsulinemia and more effective than metformin in the treatment of experimental insulin-responsive breast cancer. Alloxan 79-86 insulin like growth factor 1 Homo sapiens 0-4 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 nitric oxide synthase 2 Rattus norvegicus 82-113 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 nitric oxide synthase 2 Rattus norvegicus 115-119 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 tumor necrosis factor Rattus norvegicus 263-290 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 tumor necrosis factor Rattus norvegicus 292-301 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 interleukin 1 beta Rattus norvegicus 304-321 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 interleukin 1 beta Rattus norvegicus 323-331 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 interleukin 6 Rattus norvegicus 338-351 22129064-4 2012 RHSE inhibited alloxan-induced nitric oxide (NO) generation through inhibition of inducible nitric oxide synthase (iNOS) gene expression and suppressed the inflammatory reaction in INS-1 cells through inhibition of expression of pro-inflammatory genes, including tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and interleukin-6 (IL-6). Alloxan 15-22 interleukin 6 Rattus norvegicus 353-357 22785400-5 2012 After 15 weeks, alloxan-treated diabetic rabbits with expected high blood glucose showed ~5-fold increase in Abeta40/Abeta42 in cortex and hippocampus, and significantly, generated Abeta-derived assemblies found in human AD. Alloxan 16-23 amyloid beta precursor protein Homo sapiens 109-114 21969757-4 2010 The erythrocytes membrane lipid peroxide and catalase activity was increased where as the activities of superoxide dismutase, glutathione peroxidase were found to be decreased significantly (P<0.01) in alloxan-induced diabetic rats. Alloxan 205-212 catalase Rattus norvegicus 45-53 20640461-5 2011 Here, we describe the synthesis of novel UDP-GlcNAc/UDP analogues and evaluate their inhibitory properties and structural binding modes in vitro alongside alloxan, a previously reported weak OGT inhibitor. Alloxan 155-162 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 191-194 21705841-0 2011 Berberine ameliorates hyperglycemia in alloxan-induced diabetic C57BL/6 mice through activation of Akt signaling pathway. Alloxan 39-46 thymoma viral proto-oncogene 1 Mus musculus 99-102 21998672-3 2011 In this paper, a number of alloxan-based compounds, initially conceived to bias other therapeutically relevant enzymes, were rationally modified and successfully repurposed to inhibit MMP-2 (also named gelatinase A) in the nanomolar range. Alloxan 27-34 matrix metallopeptidase 2 Homo sapiens 184-189 21689743-5 2011 In addition, we found that Alloxan induces AMPK differently from IL1beta, as Alloxan acts mainly through CaMKII while IL1beta acts through LKB1 phosphorylation. Alloxan 27-34 calcium/calmodulin-dependent protein kinase II, beta Mus musculus 105-111 21689743-5 2011 In addition, we found that Alloxan induces AMPK differently from IL1beta, as Alloxan acts mainly through CaMKII while IL1beta acts through LKB1 phosphorylation. Alloxan 27-34 interleukin 1 beta Mus musculus 118-125 21689743-5 2011 In addition, we found that Alloxan induces AMPK differently from IL1beta, as Alloxan acts mainly through CaMKII while IL1beta acts through LKB1 phosphorylation. Alloxan 27-34 serine/threonine kinase 11 Mus musculus 139-143 21689743-5 2011 In addition, we found that Alloxan induces AMPK differently from IL1beta, as Alloxan acts mainly through CaMKII while IL1beta acts through LKB1 phosphorylation. Alloxan 77-84 calcium/calmodulin-dependent protein kinase II, beta Mus musculus 105-111 21969757-5 2010 The levels of lipid peroxide in liver of diabetic rats increased significantly (P<0.01) and catalase, superoxide dismutase, glutathione peroxidase in liver was significantly decreased in alloxan-induced diabetic rats, when compared to normal rats. Alloxan 190-197 catalase Rattus norvegicus 95-103 20165912-2 2010 Alloxan, an inhibitor of O-GlcNAc transferase, potentiated responses of AMPA receptors composed of the GluR1 subunit expressed in Xenopus oocytes. Alloxan 0-7 glutamate receptor, ionotropic, AMPA 1 L homeolog Xenopus laevis 103-108 20600753-3 2010 Different works have demonstrated that NSF extract showed antihyperglycemic effect on alloxan-induced diabetic rats. Alloxan 86-93 N-ethylmaleimide sensitive factor, vesicle fusing ATPase Rattus norvegicus 39-42 20610567-7 2010 In an animal model of diabetes induced by alloxan, progression of hyperglycemia was significantly attenuated in mice given the Ad-GLP-1 vector compared with control mice. Alloxan 42-49 glucagon Mus musculus 130-135 20429050-9 2010 Our data also showed both that EMs inhibited cell apoptosis and cell cycle G1 arrest induced by STZ and ALX through down-regulaing p53 and p21 expression. Alloxan 104-107 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 131-134 20429050-9 2010 Our data also showed both that EMs inhibited cell apoptosis and cell cycle G1 arrest induced by STZ and ALX through down-regulaing p53 and p21 expression. Alloxan 104-107 KRAS proto-oncogene, GTPase Rattus norvegicus 139-142 20447389-7 2010 Our results revealed that in alloxan-induced diabetic mice, the nuclear staining of NF-kappaB p65 was increased in glomerulus, whereas renal I kappaB-alpha protein was significantly reduced. Alloxan 29-36 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 84-93 20447389-7 2010 Our results revealed that in alloxan-induced diabetic mice, the nuclear staining of NF-kappaB p65 was increased in glomerulus, whereas renal I kappaB-alpha protein was significantly reduced. Alloxan 29-36 v-rel reticuloendotheliosis viral oncogene homolog A (avian) Mus musculus 94-97 20447389-7 2010 Our results revealed that in alloxan-induced diabetic mice, the nuclear staining of NF-kappaB p65 was increased in glomerulus, whereas renal I kappaB-alpha protein was significantly reduced. Alloxan 29-36 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 141-155 20130740-3 2010 HPS3 was selected for examination of its hypoglycemic mechanism because of its significant hypoglycemic effect in alloxan-induced diabetic mice. Alloxan 114-121 HPS3, biogenesis of lysosomal organelles complex 2 subunit 1 Mus musculus 0-4 17768029-0 2009 Hepatic glucokinase activity is the primary defect in alloxan-induced diabetes of mice. Alloxan 54-61 glucokinase Mus musculus 8-19 20336201-6 2009 In vitro studies using cerebral extract from alloxan diabetic rats demonstrated significant (P<0.05) inhibition of AChE activity in the brain of normal animals. Alloxan 45-52 acetylcholinesterase Rattus norvegicus 118-122 20722995-1 2010 OBJECTIVES: In rats with diabetes mellitus induced by alloxan (DMIA) or streptozocin (DMIS), changes in the cytochrome P450 (CYP) isozymes in the liver, lung, kidney, intestine, brain, and testis have been reported based on Western blot analysis, Northern blot analysis, and various enzyme activities. Alloxan 54-61 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 108-123 20722995-1 2010 OBJECTIVES: In rats with diabetes mellitus induced by alloxan (DMIA) or streptozocin (DMIS), changes in the cytochrome P450 (CYP) isozymes in the liver, lung, kidney, intestine, brain, and testis have been reported based on Western blot analysis, Northern blot analysis, and various enzyme activities. Alloxan 54-61 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 125-128 19563792-8 2009 CONCLUSION: These results indicate that low catalase activity in the blood is associated with the diabetes mellitus caused by alloxan administration. Alloxan 126-133 catalase Mus musculus 44-52 19454312-1 2009 AIM OF THE STUDY: Total flavonids of Polygonatum(P) odoratum (TFP) were tested for anti-diabetic activity in streptozotocin (STZ)-induced diabetic mice and alloxan-induced diabetic rats. Alloxan 156-163 tripartite motif-containing 39 Mus musculus 18-60 19454312-4 2009 The experiments were designed to detect the anti-diabetic activity of TFP by determination of blood glucose (BG) using one touch gluco-meter and insulin levels by using a radioimmunoassay kit in streptozotocin (STZ)-induced diabetic mice and alloxan-induced diabetic rats and alpha-amylase inhibitory activity by alpha-amylase inhibition assay in vitro. Alloxan 242-249 tripartite motif-containing 39 Mus musculus 70-73 19454312-10 2009 TFP also could increase significantly the insulin level in alloxan-induced type 2 diabetic rats (P<0.05) compared with control. Alloxan 59-66 tripartite motif-containing 39 Mus musculus 0-3 19268552-0 2009 The plasmid encoding HSP47 enhances collagen expression and promotes skin wound healing in an alloxan-induced diabetic model. Alloxan 94-101 serine (or cysteine) peptidase inhibitor, clade H, member 1 Mus musculus 21-26 17768029-5 2009 Alloxan is believed to confer its diabetogenic effect by inhibiting pancreatic glucokinase activity, leading to pancreatic beta-cell death. Alloxan 0-7 glucokinase Mus musculus 79-90 17768029-7 2009 Our results show that alloxan treatment led to an 81% reduction in glucokinase immunoreactivity and a greater than 90% reduction in glucokinase enzymatic activity in the liver, suggesting that alloxan"s toxicity is not specific to the pancreas. Alloxan 22-29 glucokinase Mus musculus 67-78 17768029-7 2009 Our results show that alloxan treatment led to an 81% reduction in glucokinase immunoreactivity and a greater than 90% reduction in glucokinase enzymatic activity in the liver, suggesting that alloxan"s toxicity is not specific to the pancreas. Alloxan 22-29 glucokinase Mus musculus 132-143 18949387-8 2008 Of note, alloxan, an O-N-acetylglucosamine transferase inhibitor, which prevented the glucosamine-induced O-GlcNAc modification, abrogated the suppressive effect of glucosamine on MCP-1 and ICAM-1 expression (p<0.05 at 0.5 mM). Alloxan 9-16 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 106-114 20830981-0 2009 Catalase activity, lipid peroxidation, cholesterol and triglyceride levels in alloxan--induced diabetes mellitus in female and male rats. Alloxan 78-85 catalase Rattus norvegicus 0-8 18711746-3 2009 The current study showed that alloxan-induced diabetic animals revealed hyperglycemia and was associated with an increase in the content of 5-HT, PKC-alpha expression and PKC activity (P < 0.05) simultaneously in striatum (ST), midbrain (MB), pons medulla (PM), cerebellum (CB), and cerebral cortex (CCX) from 7 days to 60 days. Alloxan 30-37 protein kinase C, alpha Rattus norvegicus 146-155 18711746-3 2009 The current study showed that alloxan-induced diabetic animals revealed hyperglycemia and was associated with an increase in the content of 5-HT, PKC-alpha expression and PKC activity (P < 0.05) simultaneously in striatum (ST), midbrain (MB), pons medulla (PM), cerebellum (CB), and cerebral cortex (CCX) from 7 days to 60 days. Alloxan 30-37 protein kinase C, alpha Rattus norvegicus 146-149 19120287-4 2008 The selection of ALR, for resistance against alloxan-induced free radical-mediated diabetes, led to a strain of mice with an elevated systemic as well as pancreatic ROS dissipation. Alloxan 45-52 growth factor, augmenter of liver regeneration Mus musculus 17-20 18949387-8 2008 Of note, alloxan, an O-N-acetylglucosamine transferase inhibitor, which prevented the glucosamine-induced O-GlcNAc modification, abrogated the suppressive effect of glucosamine on MCP-1 and ICAM-1 expression (p<0.05 at 0.5 mM). Alloxan 9-16 C-C motif chemokine ligand 2 Homo sapiens 180-185 18949387-8 2008 Of note, alloxan, an O-N-acetylglucosamine transferase inhibitor, which prevented the glucosamine-induced O-GlcNAc modification, abrogated the suppressive effect of glucosamine on MCP-1 and ICAM-1 expression (p<0.05 at 0.5 mM). Alloxan 9-16 intercellular adhesion molecule 1 Homo sapiens 190-196 19004374-2 2008 Pigs were made diabetic with single dose of alloxan, which acts by selectively destroying insulin-producing pancreatic beta cells thus inducing type 1 diabetes. Alloxan 44-51 insulin Sus scrofa 90-97 18478316-1 2008 PURPOSE: It has been reported that telithromycin is primarily metabolized via hepatic CYP3A1/2 in rats, the expression and/or mRNA level of hepatic CYP3A1/2 increase in rat model of diabetes mellitus induced by alloxan (DMIA) or streptozotocin (DMIS), and intestinal CYP3A1/2 enzyme activity decreases in rat model of DMIS. Alloxan 211-218 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 148-154 18758143-8 2008 Furthermore, although oral administration of scutellarin (10 and 50 mg/kg) did not produce significant antihyperglycemic action, it lowered the serum MCP-1 levels significantly in alloxan-induced diabetic mice. Alloxan 180-187 chemokine (C-C motif) ligand 2 Mus musculus 150-155 18690293-0 2008 PPAR gamma agonist normalizes glomerular filtration rate, tissue levels of homocysteine, and attenuates endothelial-myocyte uncoupling in alloxan induced diabetic mice. Alloxan 138-145 peroxisome proliferator activated receptor gamma Mus musculus 0-10 18478316-1 2008 PURPOSE: It has been reported that telithromycin is primarily metabolized via hepatic CYP3A1/2 in rats, the expression and/or mRNA level of hepatic CYP3A1/2 increase in rat model of diabetes mellitus induced by alloxan (DMIA) or streptozotocin (DMIS), and intestinal CYP3A1/2 enzyme activity decreases in rat model of DMIS. Alloxan 211-218 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 148-154 18442208-0 2008 Ghrelin improves delayed gastrointestinal transit in alloxan-induced diabetic mice. Alloxan 53-60 ghrelin Mus musculus 0-7 19024533-9 2008 The relative values of nNOS mRNA expression 15, 30, and 45 days after alloxan injection of diabetic ED rabbits were 0.670 +/- 0.030, 0.451 +/- 0.012, and 0.206 +/- 0.023 respectively,all significantly lower than those of the control group (0.817 +/- 0.010, 0.814 +/- 0.020, and 0.802 +/- 0.007 respectively, all P < 0.05). Alloxan 70-77 nitric oxide synthase, brain Oryctolagus cuniculus 23-27 19024533-10 2008 Western blotting showed that the relative values of nNOS 15, 30, and 45 days after alloxan injection of the ED rabbits were 0.713 +/- 0.014, 0.424 +/- 0.007, and 0.337 +/- 0.009 respectively, all significantly lower than those of the control group (0.797 +/- 0.015, 0.706 +/- 0.020, and 0. Alloxan 83-90 nitric oxide synthase, brain Oryctolagus cuniculus 52-56 18516449-2 2008 METHODS: evaluations were performed in three groups: A--8 weeks of age, B--44 weeks of age, C--44 weeks of age with alloxan-induced diabetes. Alloxan 116-123 alpha-1-B glycoprotein Rattus norvegicus 92-97 18087688-1 2008 Alloxan and streptozotocin are toxic glucose analogues that preferentially accumulate in pancreatic beta cells via the GLUT2 glucose transporter. Alloxan 0-7 solute carrier family 2 member 2 Homo sapiens 119-124 17116429-8 2007 Furthermore, UNG-proteins and SMUG1 may have important functions in removal of oxidized cytosines, e.g. isodialuric acid, alloxan and 5-hydroxyuracil after exposure to ionizing radiation. Alloxan 122-129 uracil DNA glycosylase Mus musculus 13-16 18212479-4 2008 Western blot and immunohistochemical analyses revealed that BPRP levels were decreased in the hippocampal CA1 neurons of diabetic rats 4 and 8 weeks post-alloxan injection and in PC12 cells 48 h after exposure to high concentrations of glucose. Alloxan 154-161 carbonic anhydrase 1 Rattus norvegicus 106-109 18034661-8 2007 HSE at 200 mg/kg attenuated the alloxan-induced decrease in the activities of superoxide dismutase (SOD), catalase (CAT) and the level of glutathione (GSH) by 36%, 44%, and 64% in the liver and by 20%, 43%, and 85% in the kidney of rats. Alloxan 32-39 catalase Rattus norvegicus 106-114 18034661-8 2007 HSE at 200 mg/kg attenuated the alloxan-induced decrease in the activities of superoxide dismutase (SOD), catalase (CAT) and the level of glutathione (GSH) by 36%, 44%, and 64% in the liver and by 20%, 43%, and 85% in the kidney of rats. Alloxan 32-39 catalase Rattus norvegicus 116-119 17101234-4 2007 Although uracil is the main substrate of uracil-DNA glycosylases UNG1 and UNG2, these proteins also remove the oxidized cytosine derivatives isodialuric acid, alloxan and 5-hydroxyuracil. Alloxan 159-166 uracil DNA glycosylase Homo sapiens 65-69 17101234-4 2007 Although uracil is the main substrate of uracil-DNA glycosylases UNG1 and UNG2, these proteins also remove the oxidized cytosine derivatives isodialuric acid, alloxan and 5-hydroxyuracil. Alloxan 159-166 uracil DNA glycosylase Homo sapiens 74-78 17658282-6 2007 Under normal physiological conditions, or during diabetes induction with alloxan or multiple low doses of streptozotocin, blood glucose was significantly lower in mice overexpressing endothelial nitric oxide synthase compared to wildtype or knockout mice. Alloxan 73-80 nitric oxide synthase 3, endothelial cell Mus musculus 183-216 17116429-8 2007 Furthermore, UNG-proteins and SMUG1 may have important functions in removal of oxidized cytosines, e.g. isodialuric acid, alloxan and 5-hydroxyuracil after exposure to ionizing radiation. Alloxan 122-129 single-strand-selective monofunctional uracil-DNA glycosylase 1 Homo sapiens 30-35 18806305-3 2007 While a single dose of alloxan (120 mg/kg) increased the serum levels of glucose and alpha-amylase activity, rate of water consumption and lipid peroxidation (LPO) in hepatic, cardiac and renal tissues with a parallel decrease in serum insulin level, administration of 25 mg/kg of CS or 200 mg/kg of PG was found to normalize all the adverse changes induced by alloxan, revealing the antidiabetic and anti peroxidative potential of test fruit peel extracts. Alloxan 23-30 alpha-amylase Citrus sinensis 85-98 17069847-8 2007 O-GlcNAc transferase (OGT) catalyzes the formation of O-GlcNAc, and inhibition of OGT with 5 mM alloxan also reversed the protection associated with glutamine. Alloxan 96-103 O-linked N-acetylglucosamine (GlcNAc) transferase Rattus norvegicus 0-20 17867477-0 2007 Effect of a compound recipe (medicinal plants) on serum insulin levels of alloxan induced diabetic rabbits. Alloxan 74-81 insulin Oryctolagus cuniculus 56-63 17867477-10 2007 CONCLUSION: From this study it may be concluded that the Compound recipe causes an increase in serum insulin levels in alloxan induced diabetic rabbits possibly due to regeneration of pancreatic beta cells. Alloxan 119-126 insulin Oryctolagus cuniculus 101-108 17069847-8 2007 O-GlcNAc transferase (OGT) catalyzes the formation of O-GlcNAc, and inhibition of OGT with 5 mM alloxan also reversed the protection associated with glutamine. Alloxan 96-103 O-linked N-acetylglucosamine (GlcNAc) transferase Rattus norvegicus 22-25 17069847-8 2007 O-GlcNAc transferase (OGT) catalyzes the formation of O-GlcNAc, and inhibition of OGT with 5 mM alloxan also reversed the protection associated with glutamine. Alloxan 96-103 O-linked N-acetylglucosamine (GlcNAc) transferase Rattus norvegicus 82-85 17045574-0 2006 Alloxan is an inhibitor of O-GlcNAc-selective N-acetyl-beta-D-glucosaminidase. Alloxan 0-7 O-GlcNAcase Homo sapiens 27-77 17146556-6 2006 Further characterization of this molecule by reverse-phase hydrophobic HPLC chromatographic analysis as well as its antidiabetic activity on alloxan-induced mice showed that it has insulin-like properties. Alloxan 141-148 insulin Sus scrofa 181-188 17045574-9 2006 Together, these data demonstrate that alloxan is an inhibitor of O-GlcNAc-selective N-acetyl-beta-d-glucosaminidase, with inhibition corresponding to an altered tryptic digest pattern of N-terminal active site peptides. Alloxan 38-45 O-GlcNAcase Homo sapiens 65-115 16753349-5 2006 For intravenous insulin/glucose infusion test, high-fat diet-fed and low-dose alloxan-treated Wistar rats were associated with insulin resistance, which was improved after AC or fenofibrate treatment. Alloxan 78-85 insulin Homo sapiens 16-23 16895803-1 2006 The diabetogenic agent alloxan is selectively accumulated in insulin-producing cells through uptake via the GLUT2 glucose transporter in the plasma membrane. Alloxan 23-30 insulin Homo sapiens 61-68 16895803-1 2006 The diabetogenic agent alloxan is selectively accumulated in insulin-producing cells through uptake via the GLUT2 glucose transporter in the plasma membrane. Alloxan 23-30 solute carrier family 2 member 2 Homo sapiens 108-113 16753349-5 2006 For intravenous insulin/glucose infusion test, high-fat diet-fed and low-dose alloxan-treated Wistar rats were associated with insulin resistance, which was improved after AC or fenofibrate treatment. Alloxan 78-85 insulin Homo sapiens 127-134 16716913-12 2006 daily for 7 days, CSP-1 produced a significant drop in blood glucose level in both STZ-induced diabetic rats and alloxan-induced diabetic mice. Alloxan 113-120 common salivary protein 1 Rattus norvegicus 18-23 16835866-4 2006 In addition, alloxan induced a notable increase in the expression of CD8+ lymphocytes to form a dramatic decrease in CD4+/CD8+ ratio (while CD4+ was unchanged). Alloxan 13-20 CD4 antigen Mus musculus 117-120 16567528-9 2006 The alloxan-induced approximately 60% deficit in beta-cell mass lead to an approximately 70% deficit in postprandial insulin secretion and loss of the postprandial insulin-driven suppression of glucagon secretion. Alloxan 4-11 insulin Sus scrofa 117-124 16516154-3 2006 In the present study, we therefore investigated apoM expression and secretion in NMRI mice rendered diabetes through administration of alloxan (120 mg/kg). Alloxan 135-142 apolipoprotein M Mus musculus 48-52 14985051-1 2004 The influence of alloxan-induced diabetes on the reactivity of rabbit basilar artery to endothelin-1 was examined. Alloxan 17-24 endothelin-1 Oryctolagus cuniculus 88-100 16684669-0 2006 Interleukin-6 and oxidative stress in plasma of alloxan-induced diabetic rabbits after pioglitazone treatment. Alloxan 48-55 interleukin-6 Oryctolagus cuniculus 0-13 15277373-6 2004 The glucokinase inhibitor alloxan increases KATP single-channel currents in glucose-excited neurons in a manner similar to low glucose. Alloxan 26-33 glucokinase Rattus norvegicus 4-15 15226592-4 2004 Our approach was to determine the effect of injecting alloxan, a GK inhibitor, into the 4V on pulsatile luteinizing hormone (LH) secretion. Alloxan 54-61 glucokinase Rattus norvegicus 65-67 15652272-3 2005 flowers (ACF) was screened for its antioxidant effect in alloxan induced diabetic rats. Alloxan 57-64 APOBEC1 complementation factor Rattus norvegicus 9-12 14758569-2 2004 Here, we describe differential in vitro effects of STZ and ALX on beta-cell molecules that are essential for glucose transport and metabolism, the glucose transporter 2 (GLUT2) and glucokinase (GK), respectively. Alloxan 59-62 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 147-168 14666367-0 2004 Combined gastrin and epidermal growth factor treatment induces islet regeneration and restores normoglycaemia in C57Bl6/J mice treated with alloxan. Alloxan 140-147 gastrin Mus musculus 9-16 14666367-0 2004 Combined gastrin and epidermal growth factor treatment induces islet regeneration and restores normoglycaemia in C57Bl6/J mice treated with alloxan. Alloxan 140-147 epidermal growth factor Mus musculus 21-44 14758569-2 2004 Here, we describe differential in vitro effects of STZ and ALX on beta-cell molecules that are essential for glucose transport and metabolism, the glucose transporter 2 (GLUT2) and glucokinase (GK), respectively. Alloxan 59-62 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 170-175 14758569-2 2004 Here, we describe differential in vitro effects of STZ and ALX on beta-cell molecules that are essential for glucose transport and metabolism, the glucose transporter 2 (GLUT2) and glucokinase (GK), respectively. Alloxan 59-62 glucokinase Mus musculus 181-192 14758569-2 2004 Here, we describe differential in vitro effects of STZ and ALX on beta-cell molecules that are essential for glucose transport and metabolism, the glucose transporter 2 (GLUT2) and glucokinase (GK), respectively. Alloxan 59-62 glucokinase Mus musculus 194-196 14758569-4 2004 ALX concentration-dependently reduced the mRNA expression of GLUT2 and GK and the effect on GLUT2 was more marked. Alloxan 0-3 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 61-66 14758569-4 2004 ALX concentration-dependently reduced the mRNA expression of GLUT2 and GK and the effect on GLUT2 was more marked. Alloxan 0-3 glucokinase Mus musculus 71-73 14758569-4 2004 ALX concentration-dependently reduced the mRNA expression of GLUT2 and GK and the effect on GLUT2 was more marked. Alloxan 0-3 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 92-97 14758569-11 2004 Preincubation with D-G, however, protected the cultures from ALX-induced reduction of GLUT2 and GK mRNA expression, whereas 5-T-G, at best, exerted a modest protection against ALX at a concentration of 1 mmol/l. Alloxan 61-64 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 86-91 14758569-11 2004 Preincubation with D-G, however, protected the cultures from ALX-induced reduction of GLUT2 and GK mRNA expression, whereas 5-T-G, at best, exerted a modest protection against ALX at a concentration of 1 mmol/l. Alloxan 61-64 glucokinase Mus musculus 96-98 14614541-7 2003 Injection in alloxan-treated rats with purified recombinant fusion adiponectin or gAdiponectin transiently abolished hyperglycemia. Alloxan 13-20 adiponectin, C1Q and collagen domain containing Rattus norvegicus 67-78 12625506-6 2003 Acute toxic effects of alloxan (hypoglycemia, hyperglycemia, nephrotoxicosis) were minimized by preventive fluid loading and by use of algorithms in which insulin, food, and fluid therapy were administered. Alloxan 23-30 insulin Sus scrofa 155-162 12928773-6 2003 RESULTS: MMP-9 activity, measured by zymography, increased in plasma and in the left ventricle of alloxan-induced diabetic wild-type mice. Alloxan 98-105 matrix metallopeptidase 9 Mus musculus 9-14 14552552-3 2003 Preceding anxiogenic stress increased the extent of the alloxan-induced increase in cerebral MAO-B activity and the accompanying abnormalities in the rats" behavior, and also potentiated the hyperglycemic effect of alloxan. Alloxan 56-63 monoamine oxidase B Rattus norvegicus 93-98 12605241-1 2003 Corpus cavernosum smooth muscle (CCSM) from rabbits made diabetic for 6 months as a result of alloxan injection exhibited increased sensitivity (3vs 9 nM EC(50)) and generated 20-50% greater force to endothelin-1 (ET-1) compared to CCSM from normal rabbits. Alloxan 94-101 endothelin-1 Oryctolagus cuniculus 200-212 12605241-1 2003 Corpus cavernosum smooth muscle (CCSM) from rabbits made diabetic for 6 months as a result of alloxan injection exhibited increased sensitivity (3vs 9 nM EC(50)) and generated 20-50% greater force to endothelin-1 (ET-1) compared to CCSM from normal rabbits. Alloxan 94-101 endothelin-1 Oryctolagus cuniculus 214-218 12625507-11 2003 We conclude that hyperglycemia is a major cause of insulin resistance in the porcine model of alloxan-induced diabetes mellitus and dyslipidemia. Alloxan 94-101 insulin Sus scrofa 51-58 14598126-6 2003 With the use of the subcutaneous insulin lispro infusion algorithm, which we have previously reported, alloxan-induced diabetic dogs exhibited postprandial hyperglycemia and delayed hyperinsulinemia, followed by hypoglycemia after an oral glucose load of 2 g/kg. Alloxan 103-110 insulin Canis lupus familiaris 33-40 12054585-4 2002 In isolated pancreatic islets, alloxan almost completely blocked both glucosamine-induced and STZ-induced protein O-GlcNAcylation, suggesting that alloxan indeed was inhibiting (OGT). Alloxan 31-38 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 178-181 12413707-4 2002 The extracts reduced blood glucose level 2 h following administration in both normal and alloxan-induced diabetic mice. Alloxan 89-96 blood glucose level 2 Mus musculus 21-42 12137914-6 2002 Ex vivo, a gradual decrement of both GLUT2 and GK mRNA expression was found in islets isolated from ALX-treated C57BL/6 mice. Alloxan 100-103 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 37-42 12137914-6 2002 Ex vivo, a gradual decrement of both GLUT2 and GK mRNA expression was found in islets isolated from ALX-treated C57BL/6 mice. Alloxan 100-103 glucokinase Mus musculus 47-49 12137914-9 2002 Pretreatment with D-glucose (D-G) protected the mRNA expression of GLUT2 and GK against ALX toxicity and prevented diabetes. Alloxan 88-91 solute carrier family 2 (facilitated glucose transporter), member 2 Mus musculus 67-72 12137914-9 2002 Pretreatment with D-glucose (D-G) protected the mRNA expression of GLUT2 and GK against ALX toxicity and prevented diabetes. Alloxan 88-91 glucokinase Mus musculus 77-79 12436338-1 2002 AIMS/HYPOTHESIS: We investigated the importance of the low affinity GLUT2 glucose transporter in the diabetogenic action of alloxan in bioengineered RINm5F insulin-producing cells with different expressions of the transporter. Alloxan 124-131 solute carrier family 2 member 2 Rattus norvegicus 68-73 12054585-4 2002 In isolated pancreatic islets, alloxan almost completely blocked both glucosamine-induced and STZ-induced protein O-GlcNAcylation, suggesting that alloxan indeed was inhibiting (OGT). Alloxan 147-154 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 178-181 12054585-5 2002 In order to show definitively that alloxan was inhibiting OGT activity, recombinant OGT was incubated with 0-10 mM alloxan, and OGT activity was measured directly by quantitating UDP-[(3)H]-GlcNAc incorporation into the recombinant protein substrate, nucleoporin p62. Alloxan 35-42 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 58-61 12054585-5 2002 In order to show definitively that alloxan was inhibiting OGT activity, recombinant OGT was incubated with 0-10 mM alloxan, and OGT activity was measured directly by quantitating UDP-[(3)H]-GlcNAc incorporation into the recombinant protein substrate, nucleoporin p62. Alloxan 35-42 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 84-87 12054585-5 2002 In order to show definitively that alloxan was inhibiting OGT activity, recombinant OGT was incubated with 0-10 mM alloxan, and OGT activity was measured directly by quantitating UDP-[(3)H]-GlcNAc incorporation into the recombinant protein substrate, nucleoporin p62. Alloxan 35-42 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 84-87 12054585-5 2002 In order to show definitively that alloxan was inhibiting OGT activity, recombinant OGT was incubated with 0-10 mM alloxan, and OGT activity was measured directly by quantitating UDP-[(3)H]-GlcNAc incorporation into the recombinant protein substrate, nucleoporin p62. Alloxan 35-42 nucleoporin 62 Homo sapiens 263-266 12054585-6 2002 Under these conditions, OGT activity was completely inhibited by 1 mM alloxan with half-maximal inhibition achieved at a concentration of 0.1 mM alloxan. Alloxan 70-77 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 24-27 12054585-6 2002 Under these conditions, OGT activity was completely inhibited by 1 mM alloxan with half-maximal inhibition achieved at a concentration of 0.1 mM alloxan. Alloxan 145-152 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 24-27 12054585-7 2002 Together, these data demonstrate that alloxan is an inhibitor of OGT, and as such, is the first OGT inhibitor described. Alloxan 38-45 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 65-68 12054585-7 2002 Together, these data demonstrate that alloxan is an inhibitor of OGT, and as such, is the first OGT inhibitor described. Alloxan 38-45 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 96-99 11817013-4 2001 Alloxan treatment is associated with a significant increase of thiobarbituric acid reactive substances and the activity of antioxidant enzymes superoxide dismutase and catalase. Alloxan 0-7 catalase Rattus norvegicus 168-176 11522665-10 2001 After alloxan administration, insulin secretion and insulin pulse mass (but not insulin pulse interval) decreased in relation to beta-cell mass. Alloxan 6-13 insulin Homo sapiens 30-37 11522665-13 2001 We conclude that an alloxan-induced selective decrease in beta-cell mass leads to deficient insulin secretion by attenuating insulin pulse mass, and that the latter is associated with decreased hepatic insulin clearance and relative hyperglucagonemia, thereby emulating the pattern of islet dysfunction observed in type 2 diabetes. Alloxan 20-27 insulin Homo sapiens 92-99 11460883-6 2001 NFkappaB activation in the pancreas 30 min after alloxan injection (60 mg/kg, iv), as assessed by an electrophoretic mobility shift assay, was not detected in the mice pretreated with A. Alloxan 49-56 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 0-8 11510477-4 2001 The formation of DNA ladder was prevented by vitamin E, BHA and catalase, suggesting that the ROS is involved in the process of apoptosis in INS-1 cells treated with alloxan. Alloxan 166-173 catalase Rattus norvegicus 45-72 10751203-0 2000 Islet amyloid polypeptide (amylin)-deficient mice develop a more severe form of alloxan-induced diabetes. Alloxan 80-87 islet amyloid polypeptide Mus musculus 27-33 10902577-7 2000 Injection of NO in alloxan-induced diabetic mice mimicked the effect of insulin in the control of hyperglycemia (i.e., lowered the glucose content in plasma). Alloxan 19-26 insulin Homo sapiens 72-79 11790282-0 2001 Endothelial nitric oxide synthase protein expression, localization, and activity in the penis of the alloxan-induced diabetic rat. Alloxan 101-108 nitric oxide synthase 3 Rattus norvegicus 0-33 11790282-10 2001 CONCLUSIONS: In the penis of the alloxan-induced diabetic rat, eNOS protein expression and synthetic activity were reduced compared with the normal rat penis, independent of testosterone influence and in the absence of significant erectile tissue degenerative changes. Alloxan 33-40 nitric oxide synthase 3 Rattus norvegicus 63-67 10700381-4 2000 Controlled proteolysis of wild-type glucokinase by proteinase K revealed that the SH group oxidizing agent alloxan can induce the formation of multiple intramolecular disulfide bridges corresponding to a double-band pattern of glucokinase protein in nonreducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Alloxan 107-114 glucokinase Homo sapiens 36-47 10700381-4 2000 Controlled proteolysis of wild-type glucokinase by proteinase K revealed that the SH group oxidizing agent alloxan can induce the formation of multiple intramolecular disulfide bridges corresponding to a double-band pattern of glucokinase protein in nonreducing sodium dodecyl sulfate-polyacrylamide gel electrophoresis. Alloxan 107-114 glucokinase Homo sapiens 227-238 11053894-3 2000 A single dose of alloxan (100 mg/kg) produced a decrease in insulin level, hyperglycemia, elevated total lipids, triglycerides and cholesterol, decreased high-density lipoprotein and hepatic glycogen contents and elevated hepatic glucose-6-phosphatase activity. Alloxan 17-24 glucose-6-phosphatase catalytic subunit 1 Rattus norvegicus 230-251 10868954-4 2000 After a 24-h culture with IL-1beta (30 U/ml), beta-cells exhibited a lower expression of the beta-cell-specific protein transcription factor pancreatic and duodenal homeobox gene (PDX)-1, glucose transporter GLUT2, and proinsulin convertase PC2, with a marked reduction (60-70%) in glucose-induced insulin production and selective sensitivity to the toxins alloxan (ALX) and streptozotocin (STZ). Alloxan 366-369 interleukin 1 beta Rattus norvegicus 26-34 10868954-6 2000 This IL-1beta-induced alteration in beta-cell phenotype resulted in a reduced cellular sensitivity to the beta-cell-specific toxins ALX and STZ; the production of nontoxic conditions of nitric oxide (NO) also rendered the cells less susceptible to radical-induced damage. Alloxan 132-135 interleukin 1 beta Rattus norvegicus 5-13 10704171-2 2000 The purpose of this study was to determine whether adenovirus-mediated gene transfer of endothelial NO synthase (eNOS) or Cu/Zn superoxide dismutase (SOD1) improves responsiveness to acetylcholine in alloxan-induced diabetic rabbits. Alloxan 200-207 nitric oxide synthase, endothelial Oryctolagus cuniculus 113-117 10704171-2 2000 The purpose of this study was to determine whether adenovirus-mediated gene transfer of endothelial NO synthase (eNOS) or Cu/Zn superoxide dismutase (SOD1) improves responsiveness to acetylcholine in alloxan-induced diabetic rabbits. Alloxan 200-207 superoxide dismutase [Cu-Zn] Oryctolagus cuniculus 122-148 10628433-6 1999 However, alloxan prevented insulin from increasing (P < 0.001) after feeding (131.8 pmol/1) compared with control steers (442.0 pmol/l) (pooled SEM = 47.5). Alloxan 9-16 insulin Homo sapiens 27-34 10765977-6 2000 In the liver phospholipid (PL) fraction, GLA, DGLA (dihomo-GLA) and alpha-linolenic acid (ALA) were decreased in the alloxan-treated group; in the muscle PL fraction, LA, GLA and DGLA were low, whereas an increase in the saturated fatty acid content was noted. Alloxan 117-124 galactosidase, alpha Rattus norvegicus 41-44 10765977-6 2000 In the liver phospholipid (PL) fraction, GLA, DGLA (dihomo-GLA) and alpha-linolenic acid (ALA) were decreased in the alloxan-treated group; in the muscle PL fraction, LA, GLA and DGLA were low, whereas an increase in the saturated fatty acid content was noted. Alloxan 117-124 galactosidase alpha Homo sapiens 47-50 10765977-6 2000 In the liver phospholipid (PL) fraction, GLA, DGLA (dihomo-GLA) and alpha-linolenic acid (ALA) were decreased in the alloxan-treated group; in the muscle PL fraction, LA, GLA and DGLA were low, whereas an increase in the saturated fatty acid content was noted. Alloxan 117-124 galactosidase alpha Homo sapiens 47-50 10765977-7 2000 L-arginine (the precursor of nitric oxide) and sodium nitroprusside (a nitric oxide donor) treatment of alloxan-induced diabetic rats enhanced the levels of LA, GLA and DGLA. Alloxan 104-111 galactosidase, alpha Rattus norvegicus 161-164 10535453-10 1999 A dominant genetic trait from ALR/Lt controlling this unusual AL resistance was indicated by the finding that reciprocal F1 mice of both sexes were resistant to AL administration in vivo. Alloxan 62-64 growth factor, augmenter of liver regeneration Mus musculus 30-33 11467412-0 2000 Streptozotocin and alloxan-based selection improves toxin resistance of insulin-producing RINm cells. Alloxan 19-26 insulin Homo sapiens 72-79 10661714-3 1999 Alloxan elicited significant inhibition of antioxidants including superoxide dismutase, catalase and glutathione reductase activities and decreased glutathione content in testis. Alloxan 0-7 catalase Rattus norvegicus 88-96 10661714-3 1999 Alloxan elicited significant inhibition of antioxidants including superoxide dismutase, catalase and glutathione reductase activities and decreased glutathione content in testis. Alloxan 0-7 glutathione-disulfide reductase Rattus norvegicus 101-122 9703225-4 1998 Unlike alloxan, alloxan-glutathione (GSH) and dialuric acid increased lipid peroxidation, which could be explained by the decreased activity of catalase and GSH peroxidase during incubation. Alloxan 16-23 catalase Rattus norvegicus 144-152 10506589-8 1999 Activation of NFkappaB in pancreatic nuclear extracts was observed 30 min after alloxan injection, as assessed by an electrophoretic mobility shift assay. Alloxan 80-87 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 14-22 9862424-4 1998 The activities of citrate synthase, malate synthase and malate dehydrogenase detected in the peroxisomal fraction were also increased by alloxan treatment. Alloxan 137-144 citrate synthase Rattus norvegicus 18-34 9862424-4 1998 The activities of citrate synthase, malate synthase and malate dehydrogenase detected in the peroxisomal fraction were also increased by alloxan treatment. Alloxan 137-144 citramalyl-CoA lyase Rattus norvegicus 36-51 9823015-8 1998 At 6 weeks after alloxan administration, a significant depression of the Na(+)-K+ ATPase alpha 1-subunit mRNA was noted in diabetic heart. Alloxan 17-24 ATPase Na+/K+ transporting subunit alpha 1 Rattus norvegicus 73-104 9784839-1 1998 Effect of alloxan induced diabetes on the activity of adenosine deaminase (ADA) was studied in the liver, spleen, stomach and small intestine of mice at two different postnatal ages (preweaned, 15-day and postweaned, 60-day). Alloxan 10-17 adenosine deaminase Mus musculus 54-73 9784839-1 1998 Effect of alloxan induced diabetes on the activity of adenosine deaminase (ADA) was studied in the liver, spleen, stomach and small intestine of mice at two different postnatal ages (preweaned, 15-day and postweaned, 60-day). Alloxan 10-17 adenosine deaminase Mus musculus 75-78 10468221-2 1999 Alloxan-susceptible (ALS/Lt) and AL-resistant (ALR/Lt) are inbred mouse strains derived in Japan by inbreeding CD-1 (ICR) mice with concomitant selection for high or low sensitivity to a relatively low AL dose. Alloxan 33-35 growth factor, augmenter of liver regeneration Mus musculus 47-50 10468221-4 1999 Superoxide dismutase (SOD), catalase (CAT), glutathione reductase (GR), and glutathione peroxidase (GPX) activities were determined in tissues of AL-untreated ALR/Lt and ALS/Lt male mice at 7 weeks of age. Alloxan 146-148 growth factor, augmenter of liver regeneration Mus musculus 159-162 9833941-0 1998 Evidence for direct action of alloxan to induce insulin resistance at the cellular level. Alloxan 30-37 insulin Canis lupus familiaris 48-55 9833941-6 1998 During clamps, steady state arterial insulin was higher in dogs treated with alloxan (688 +/- 60 vs 502 +/- 38 pmol/l; p = 0.023) due to a 25% reduction in insulin clearance (p = 0.045). Alloxan 77-84 insulin Canis lupus familiaris 37-44 9833941-6 1998 During clamps, steady state arterial insulin was higher in dogs treated with alloxan (688 +/- 60 vs 502 +/- 38 pmol/l; p = 0.023) due to a 25% reduction in insulin clearance (p = 0.045). Alloxan 77-84 insulin Canis lupus familiaris 156-163 9833941-10 1998 In conclusion, alloxan induces insulinopenic diabetes, with glucose intolerance and insulin resistance at the target tissue level. Alloxan 15-22 insulin Canis lupus familiaris 31-38 9048894-6 1997 The human and rat glucokinase isoforms were non-competitively inhibited by the sulfhydryl group reagents alloxan and ninhydrin with Ki values in the range of 1 microM. Alloxan 105-112 glucokinase Rattus norvegicus 18-29 9741596-1 1998 Alloxan is known to induce diabetes in experimental animals through destruction of insulin-producing 3-cells of pancreas. Alloxan 0-7 insulin Homo sapiens 83-90 9467831-1 1997 The status of cytochrome P450-dependent oxidative biotransformation of aminopyrine and benzo(a)pyrene (Phase I reaction) and glutathione S-transferase (GST) catalyzed conjugation with 1-chloro-2,4-dinitrobenzene (CDNB) (Phase II reaction) was evaluated in diabetic rats sacrificed 3 weeks after alloxan treatment (2 doses of 75 mg/kg at an interval of 48 h, i.p.). Alloxan 295-302 hematopoietic prostaglandin D synthase Rattus norvegicus 125-150 9467831-1 1997 The status of cytochrome P450-dependent oxidative biotransformation of aminopyrine and benzo(a)pyrene (Phase I reaction) and glutathione S-transferase (GST) catalyzed conjugation with 1-chloro-2,4-dinitrobenzene (CDNB) (Phase II reaction) was evaluated in diabetic rats sacrificed 3 weeks after alloxan treatment (2 doses of 75 mg/kg at an interval of 48 h, i.p.). Alloxan 295-302 hematopoietic prostaglandin D synthase Rattus norvegicus 152-155 9178105-2 1997 The adenylate cyclase inhibitor, alloxan, blocked parathyroid hormone (PTH)-stimulated acid production. Alloxan 33-40 parathyroid hormone Gallus gallus 50-69 9450640-1 1997 The effect of alloxan-induced diabetes was studied on the activity of monoamine oxidase (MAO), the oxidative deaminating enzyme of monoamine neurotransmitters. Alloxan 14-21 monoamine oxidase A Rattus norvegicus 70-87 9450640-1 1997 The effect of alloxan-induced diabetes was studied on the activity of monoamine oxidase (MAO), the oxidative deaminating enzyme of monoamine neurotransmitters. Alloxan 14-21 monoamine oxidase A Rattus norvegicus 89-92 9048894-7 1997 The inhibition of glucokinase enzyme activity was reversed by dithiothreitol with an EC50 value of 9 microM for alloxan and of 50 microM for ninhydrin. Alloxan 112-119 glucokinase Homo sapiens 18-29 9048894-8 1997 D-Glucose provided protection against alloxan-induced inhibition of human and rat glucokinase isoenzymes with half-maximal effective concentrations between 11 and 16 mM. Alloxan 38-45 glucokinase Rattus norvegicus 82-93 9048894-9 1997 The enzyme inhibition by alloxan was accompanied by a change in the electrophoretic mobility with a second lower molecular 49 kDa glucokinase band which can be interpreted as a compact glucokinase molecule locked by disulfide bonds. Alloxan 25-32 glucokinase Homo sapiens 130-141 9048894-9 1997 The enzyme inhibition by alloxan was accompanied by a change in the electrophoretic mobility with a second lower molecular 49 kDa glucokinase band which can be interpreted as a compact glucokinase molecule locked by disulfide bonds. Alloxan 25-32 glucokinase Homo sapiens 185-196 9215802-0 1997 Glutathione dependent reduction of alloxan to dialuric acid catalyzed by thioltransferase (glutaredoxin): a possible role for thioltransferase in alloxan toxicity. Alloxan 35-42 glutaredoxin-1 Sus scrofa 73-89 9215802-0 1997 Glutathione dependent reduction of alloxan to dialuric acid catalyzed by thioltransferase (glutaredoxin): a possible role for thioltransferase in alloxan toxicity. Alloxan 35-42 glutaredoxin-1 Sus scrofa 91-103 9215802-0 1997 Glutathione dependent reduction of alloxan to dialuric acid catalyzed by thioltransferase (glutaredoxin): a possible role for thioltransferase in alloxan toxicity. Alloxan 35-42 glutaredoxin-1 Sus scrofa 126-142 9215802-1 1997 Recombinant pig liver thioltransferase (rPLTT) catalyzes the reduction of alloxan to dialuric acid by glutathione (GSH). Alloxan 74-81 glutaredoxin-1 Sus scrofa 22-38 9215802-5 1997 Both superoxide dismutase and catalase inhibited oxygen consumption in 1.0 mM GSH and 0.2 mM alloxan in the presence of rPLTT. Alloxan 93-100 catalase Sus scrofa 30-38 9215802-6 1997 This study suggests that thioltransferase (glutaredoxin) plays a significant role in the cytotoxicity of alloxan in vulnerable tissues. Alloxan 105-112 glutaredoxin-1 Sus scrofa 25-41 9215802-6 1997 This study suggests that thioltransferase (glutaredoxin) plays a significant role in the cytotoxicity of alloxan in vulnerable tissues. Alloxan 105-112 glutaredoxin-1 Sus scrofa 43-55 8959353-0 1996 Renal natriuretic effects of atrial natriuretic peptide in dogs with alloxan-induced acute pulmonary edema. Alloxan 69-76 natriuretic peptide A Canis lupus familiaris 29-55 8959353-11 1996 Catalase, administered intravenously as a bolus just before the alloxan infusion, prevented pulmonary edema and the associated renal changes. Alloxan 64-71 catalase Canis lupus familiaris 0-8 8960250-2 1995 Activity of glutathione S-transferase and glutathione peroxidase in the liver of alloxan induced diabetic rats. Alloxan 81-88 hematopoietic prostaglandin D synthase Rattus norvegicus 12-37 27406273-2 1996 The toxic effects are prevented by catalase added to the medium, suggesting that alloxan does not need to be taken up in order to affect cells. Alloxan 81-88 catalase Rattus norvegicus 35-43 8161942-0 1994 Effect of alloxan-induced diabetes on acetylcholinesterase activity from discrete areas of rat brain. Alloxan 10-17 acetylcholinesterase Rattus norvegicus 38-58 8852272-6 1995 In alloxan-induced diabetes metformin (Met) treatment led to an increase in insulin receptor number in liver plasma membranes (before Met: 46.50 +/- 2.69, after Met: 76.00 +/- 3.39 fmol/mg, p < 0.001) and a decrease in plasma lipid peroxidation levels compared to the non-treated group (before Met: 1.85 +/- 0.53, after Met: 1.10 +/- 0.09 nmol MDA/ml, p < 0.05). Alloxan 3-10 insulin receptor Rattus norvegicus 76-92 8159102-2 1994 In the present study, we demonstrate that the mass of HMG CoA reductase protein is increased in the small intestine of both streptozocin-induced diabetic rats (2.5-fold) and streptozocin/alloxan-induced diabetic dogs (2.4-fold). Alloxan 187-194 3-hydroxy-3-methylglutaryl-CoA reductase Rattus norvegicus 54-71 7918687-4 1994 Treatment of beta-cells with alloxan and H2O2 caused elevation of cytosolic free Ca2+, and this increase of Ca2+ was also abolished by verapamil. Alloxan 29-36 carbonic anhydrase 2 Rattus norvegicus 81-84 7918687-4 1994 Treatment of beta-cells with alloxan and H2O2 caused elevation of cytosolic free Ca2+, and this increase of Ca2+ was also abolished by verapamil. Alloxan 29-36 carbonic anhydrase 2 Rattus norvegicus 108-111 8462284-6 1993 Injection of insulin in alloxan-induced diabetic rats is able to restore almost completely the level of c-Ki-ras transcript found in insulin-induced normal rats. Alloxan 24-31 KRAS proto-oncogene, GTPase Rattus norvegicus 104-112 8350431-5 1993 RESULTS: Both insulin-treated and untreated alloxan diabetic rabbits revealed significantly decreased oxygen tensions throughout the arterial wall compared with control rabbits. Alloxan 44-51 insulin Oryctolagus cuniculus 14-21 7679153-1 1993 A possible relationship between increased aldose reductase activity and abnormal endothelium-dependent relaxation was examined in aorta from alloxan-induced diabetic rabbits. Alloxan 141-148 aldo-keto reductase family 1 member B1 Oryctolagus cuniculus 42-58 1606640-0 1992 Enzymatic generation of alloxan radicals in rat liver microsomes: possible participation of reduced nicotinamide adenine dinucleotide phosphate (NADPH)-cytochrome P-450 reductase. Alloxan 24-31 cytochrome p450 oxidoreductase Rattus norvegicus 100-178 1638581-5 1992 The alloxan group presented the following significant findings: a decrease in white blood cell and platelet counts (44.2% and 68.2% of control, respectively) at five minutes; an increase in thromboxane B2 and 6-keto-prostaglandin F1 alpha (731.6% and 476.6% of control, respectively) at 15 minutes; an increase in beta-glucuronidase (124.8% of control) at 30 minutes; and an increase in Qwl/dQl (8.84 +/- 1.82 g/g) at the end of experiment. Alloxan 4-11 glucuronidase beta Canis lupus familiaris 314-332 1581040-8 1992 Cells pretreated with desferrioxamine (Des) and superoxide dismutase (SOD) or Des, SOD and catalase (CAT) to induce partial (H2O2 formation only) or almost full protection (no ROS formation) showed about the same reactions as when cells were exposed to alloxan and cysteine without scavengers (O2-, H2O2 and OH. Alloxan 253-260 superoxide dismutase 1 Homo sapiens 48-68 1581040-8 1992 Cells pretreated with desferrioxamine (Des) and superoxide dismutase (SOD) or Des, SOD and catalase (CAT) to induce partial (H2O2 formation only) or almost full protection (no ROS formation) showed about the same reactions as when cells were exposed to alloxan and cysteine without scavengers (O2-, H2O2 and OH. Alloxan 253-260 superoxide dismutase 1 Homo sapiens 83-86 1581040-8 1992 Cells pretreated with desferrioxamine (Des) and superoxide dismutase (SOD) or Des, SOD and catalase (CAT) to induce partial (H2O2 formation only) or almost full protection (no ROS formation) showed about the same reactions as when cells were exposed to alloxan and cysteine without scavengers (O2-, H2O2 and OH. Alloxan 253-260 catalase Homo sapiens 91-99 34511503-9 2021 O-GlcNAc transferase inhibitor alloxan could suppress DHA-induced protein O-GlcNAcylation, and subsequently prevent therapeutic effect of DHA on the deficits of learning and memory as well as synaptic plasticity in hTau mice. Alloxan 31-38 microtubule associated protein tau Homo sapiens 215-219 1676625-0 1991 Purification and aminopyrine monooxygenase activity of liver microsomal cytochrome P-450 from alloxan-induced diabetic rats. Alloxan 94-101 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 72-88 2178745-1 1990 The activity of in vitro glutathione S-transferase towards 1-chloro-2,4-dinitrobenzene was examined in liver, renal cortex, and small intestine (duodenum, jejunum, ileum) after the in vivo treatment of male Wistar rats with streptozotocin or alloxan. Alloxan 242-249 hematopoietic prostaglandin D synthase Rattus norvegicus 25-50 34668190-7 2022 Alloxan and Thiamet G were used to reduce and increase global OGT glycosylation levels in C2C12 cells, respectively. Alloxan 0-7 O-linked N-acetylglucosamine (GlcNAc) transferase (UDP-N-acetylglucosamine:polypeptide-N-acetylglucosaminyl transferase) Mus musculus 62-65 1284498-3 1992 It is reported that 14 weeks after diabetes induction with alloxan the levels of substance P and methionine-enkephalin are markedly reduced throughout the intestine, while vasoactive intestinal polypeptide content is dramatically increased. Alloxan 59-66 tachykinin precursor 1 Homo sapiens 81-92 1816091-2 1991 Since a number of central neurotransmitters are also known to influence glucose levels and it is likely that CNS insulin receptors act through neurotransmitter mediation, the present study was conducted to investigate the effect of intracerebroventricularly (icv) administered insulin on rat brain dopamine (DA), noradrenaline (NA), serotonin and acetylcholine (ACh) activity in normal and alloxan-induced hyperglycaemic animals. Alloxan 390-397 insulin Homo sapiens 113-120 2205570-2 1990 The insulin level in alloxan-induced hyperglycemic animals increased significantly after treatment with VRV. Alloxan 21-28 insulin Oryctolagus cuniculus 4-11 2185463-2 1990 Half maximal inhibitory concentrations of alloxan were greater than those previously found for half maximal inhibition of pancreatic islet or liver glucokinase. Alloxan 42-49 glucokinase Rattus norvegicus 148-159 34175449-10 2021 These findings suggest that alloxan might be a better option for animal studies regarding painful diabetic neuropathy as streptozotocin directly affects nociceptive neurons, probably by modulating TRPV1 channel activation. Alloxan 28-35 transient receptor potential cation channel, subfamily V, member 1 Rattus norvegicus 197-202 34341763-0 2021 Abrus precatorius Leaf Extract Reverses Alloxan/Nicotinamide-Induced Diabetes Mellitus in Rats through Hormonal (Insulin, GLP-1, and Glucagon) and Enzymatic (alpha-Amylase/alpha-Glucosidase) Modulation. Alloxan 40-47 glucagon Rattus norvegicus 122-127 34341763-0 2021 Abrus precatorius Leaf Extract Reverses Alloxan/Nicotinamide-Induced Diabetes Mellitus in Rats through Hormonal (Insulin, GLP-1, and Glucagon) and Enzymatic (alpha-Amylase/alpha-Glucosidase) Modulation. Alloxan 40-47 glucagon Rattus norvegicus 133-141 34629354-2 2021 The diabetogenic agents, alloxan and streptozotocin, caused DNA strand breaks, which in turn activated poly(ADP-ribose) polymerase/synthetase (PARP) to deplete NAD+, thereby inhibiting islet beta-cell functions such as proinsulin synthesis and ultimately leading to beta-cell necrosis. Alloxan 25-32 poly(ADP-ribose) polymerase 1 Homo sapiens 103-141 34629354-2 2021 The diabetogenic agents, alloxan and streptozotocin, caused DNA strand breaks, which in turn activated poly(ADP-ribose) polymerase/synthetase (PARP) to deplete NAD+, thereby inhibiting islet beta-cell functions such as proinsulin synthesis and ultimately leading to beta-cell necrosis. Alloxan 25-32 poly(ADP-ribose) polymerase 1 Homo sapiens 143-147 34629354-2 2021 The diabetogenic agents, alloxan and streptozotocin, caused DNA strand breaks, which in turn activated poly(ADP-ribose) polymerase/synthetase (PARP) to deplete NAD+, thereby inhibiting islet beta-cell functions such as proinsulin synthesis and ultimately leading to beta-cell necrosis. Alloxan 25-32 insulin Homo sapiens 219-229 35190649-10 2022 AEPAS showed a marked decrease in alloxan-induced increases in FBG, TG, LDL-c, G6P, F-1, 6-BP, MDA, IL-6, TNF-alpha, and NF-kB and increased alloxan-induced decreases in liver glycogen, hexokinase, and HDL-c. Alloxan 34-41 interleukin 6 Rattus norvegicus 100-104 35308202-16 2022 Conclusion: AECPS showed efficient ameliorative actions against alloxan-induced pancreatic dysfunction, oxidative stress suppression as well as, inflammation, and apoptosis via the activation of PI3K/AKT signaling pathways. Alloxan 64-71 AKT serine/threonine kinase 1 Rattus norvegicus 200-203 35190649-10 2022 AEPAS showed a marked decrease in alloxan-induced increases in FBG, TG, LDL-c, G6P, F-1, 6-BP, MDA, IL-6, TNF-alpha, and NF-kB and increased alloxan-induced decreases in liver glycogen, hexokinase, and HDL-c. Alloxan 34-41 tumor necrosis factor Rattus norvegicus 106-115 35190649-10 2022 AEPAS showed a marked decrease in alloxan-induced increases in FBG, TG, LDL-c, G6P, F-1, 6-BP, MDA, IL-6, TNF-alpha, and NF-kB and increased alloxan-induced decreases in liver glycogen, hexokinase, and HDL-c. Alloxan 34-41 RELA proto-oncogene, NF-kB subunit Rattus norvegicus 121-126 2745981-4 1989 Oxidant stress, in the form of alloxan or divicine, also enhanced serum levels of TNF in mice made sensitive to LPS by low-level infection with malaria, and then given i.v. Alloxan 31-38 tumor necrosis factor Mus musculus 82-85 34986201-8 2022 Alloxan-diabetic rabbits had lower levels of SOD, CAT, Gpx, and rGSH than control rabbits. Alloxan 0-7 catalase Oryctolagus cuniculus 50-53 2471969-4 1989 Injection of alloxan (125 mg/kg) induced a similar change of pancreatic enzyme pattern, with amylase activity strongly reduced by 79% and activity of lipase and colipase increased 20.5 and 18.6%, respectively. Alloxan 13-20 lipase G, endothelial type Rattus norvegicus 150-156 2536542-7 1989 In the presence of SOD, alloxan was reduced by GSH, but increasing concentrations of GSH progressively inhibited redox cycling as shown by decreased rates of O2 uptake and GSH oxidation. Alloxan 24-31 superoxide dismutase 1 Homo sapiens 19-22 2471969-4 1989 Injection of alloxan (125 mg/kg) induced a similar change of pancreatic enzyme pattern, with amylase activity strongly reduced by 79% and activity of lipase and colipase increased 20.5 and 18.6%, respectively. Alloxan 13-20 colipase Rattus norvegicus 161-169 2457529-1 1988 Exocrine secretory function in response to 10 pM to 10 nM synthetic secretin was evaluated in perfused pancreas isolated from control, streptozocin-induced diabetic (STZ-D), alloxan-induced diabetic (ALX-D), and insulin-treated STZ-D rats. Alloxan 174-181 secretin Rattus norvegicus 68-76 3207996-0 1988 Structural requirements of alloxan and ninhydrin for glucokinase inhibition and of glucose for protection against inhibition. Alloxan 27-34 glucokinase Homo sapiens 53-64 3207996-4 1988 With a half-maximal inhibitory concentration of 5 microM, alloxan was the most potent pyrimidine derivative inhibitor of glucokinase. Alloxan 58-65 glucokinase Homo sapiens 121-132 3207996-12 1988 Only glucose derivatives with a sufficiently bulky substituent in position C-2, such as the glucokinase substrates glucose and mannose and the inhibitors mannoheptulose, glucosamine, and N-acetylglucosamine, protected glucokinase against inhibition by alloxan by binding to the active site of the enzyme. Alloxan 252-259 glucokinase Homo sapiens 92-103 3207996-12 1988 Only glucose derivatives with a sufficiently bulky substituent in position C-2, such as the glucokinase substrates glucose and mannose and the inhibitors mannoheptulose, glucosamine, and N-acetylglucosamine, protected glucokinase against inhibition by alloxan by binding to the active site of the enzyme. Alloxan 252-259 glucokinase Homo sapiens 218-229 3207996-15 1988 DTT (dithiothreitol) protected glucokinase against inhibition by alloxan and reversed the inhibition of the enzyme induced by alloxan. Alloxan 65-72 glucokinase Homo sapiens 31-42 3207996-16 1988 Thus the mechanism of glucokinase inhibition by alloxan and other inhibitors, such as uramil and ninhydrin, is an oxidation of functionally essential SH groups of the enzyme, where the most reactive keto group of the inhibitor acts as the hydrogen acceptor. Alloxan 48-55 glucokinase Homo sapiens 22-33 2968936-1 1988 A common mechanism has been proposed for the beta-cell toxins alloxan (ALX) and streptozocin (STZ) involving the formation of single-strand breaks in DNA that lead to the overactivation of the enzyme poly(ADP-ribose) synthetase and the critical depletion of its substrate NAD. Alloxan 71-74 poly(ADP-ribose) polymerase 1 Homo sapiens 200-227 3419426-0 1988 Inhibition of glucokinase by alloxan through interaction with SH groups in the sugar-binding site of the enzyme. Alloxan 29-36 glucokinase Homo sapiens 14-25 3419426-2 1988 The dithiol 1,4-dithiothreitol (1,4-DTT) protected against and reversed the inhibition of glucokinase by alloxan. Alloxan 105-112 glucokinase Homo sapiens 90-101 3077897-1 1988 After intravenous administration of alloxan (50 mg kg-1 liveweight) to lactating ewes, there were triphasic changes in plasma glucose and insulin. Alloxan 36-43 LOC105613195 Ovis aries 138-145 3419426-4 1988 Only 1,3-dimercaptopropane, 1,4-dimercaptobutane, 1,4-dithioerythritol, and 1,4-DTT, with intermediate spacing between the SH groups, reversed the inhibition of glucokinase induced by alloxan. Alloxan 184-191 glucokinase Homo sapiens 161-172 3419426-8 1988 Like alloxan, other dithiol reagents such as ninhydrin, N-ethylmaleimide, and maleimide inhibited glucokinase. Alloxan 5-12 glucokinase Homo sapiens 98-109 2465668-8 1988 In rats made diabetic with alloxan or streptozotocin, the B cells were irreversibly damaged and lost both their insulin and IGF-I immunoreactivities, while the IGF-I immunoreactivity was increased in A cells; the D cells remained unchanged. Alloxan 27-34 insulin-like growth factor 1 Rattus norvegicus 124-129 2465668-8 1988 In rats made diabetic with alloxan or streptozotocin, the B cells were irreversibly damaged and lost both their insulin and IGF-I immunoreactivities, while the IGF-I immunoreactivity was increased in A cells; the D cells remained unchanged. Alloxan 27-34 insulin-like growth factor 1 Rattus norvegicus 160-165 16414626-2 1988 Diabetiogenic effect of Alloxan was utilized which produced an Insulin-Dependent Diabetic State in the animals. Alloxan 24-31 insulin Homo sapiens 63-70 3078228-2 1988 In one study it was found that the diabetes induced by alloxan could be stabilized with exogenous insulin (1.2-1.3 U h-1). Alloxan 55-62 insulin Bos taurus 98-105 3298618-1 1987 A combined pharmacokinetic/pharmacodynamic model was proposed to describe the pharmacokinetics of intravenously administered regular insulin (0.55 units/kg) in alloxan-induced diabetic dogs. Alloxan 160-167 insulin Canis lupus familiaris 133-140 3153476-1 1987 Cytochrome P450j, an enzyme involved in nitrosamine metabolism, is expressed in hepatic, pulmonary, and renal tissues and its level is elevated in ethanol- and acetone-treated rats as well as in diabetic rats induced by either streptozotocin or alloxan. Alloxan 245-252 cytochrome P450, family 2, subfamily e, polypeptide 1 Rattus norvegicus 0-16 3296598-5 1987 Alloxan inhibited glucokinase but not hexokinase activity in cytoplasmic fractions of pancreatic B-cells and liver. Alloxan 0-7 glucokinase Rattus norvegicus 18-29 3296598-7 1987 Glucokinase activity was protected from alloxan toxicity only by D-glucose and D-mannose; the alpha anomer of D-glucose provided significantly greater protection than the beta anomer. Alloxan 40-47 glucokinase Rattus norvegicus 0-11 3301234-2 1987 Alloxan induced maternal diabetes (n = 5) associated with fetal hyperglycemia and mild hyperinsulinemia (59.80 +/- 8.10 microU/ml versus a control of 26.25 +/- 3.70, p less than 0.01) increased the insulin receptor number from a control (30 d) of 168 +/- 1.01 to 320 +/- 34 X 10(10)/mg protein (p less than 0.01). Alloxan 0-7 insulin receptor Oryctolagus cuniculus 198-214 3829189-0 1986 Inhibition of rat liver glucokinase by alloxan and ninhydrin. Alloxan 39-46 glucokinase Rattus norvegicus 24-35 3427902-4 1987 A comparison of cholesterol synthesis and utilization in alloxan-induced diabetic rats and rabbits revealed that interspecies differences existed only in the response of the key enzymes regulating cholesterol utilization, namely cholesterol 7 alpha-hydroxylase and ACAT in the liver and intestine respectively. Alloxan 57-64 cytochrome P450 7A1 Oryctolagus cuniculus 229-260 3427902-4 1987 A comparison of cholesterol synthesis and utilization in alloxan-induced diabetic rats and rabbits revealed that interspecies differences existed only in the response of the key enzymes regulating cholesterol utilization, namely cholesterol 7 alpha-hydroxylase and ACAT in the liver and intestine respectively. Alloxan 57-64 sterol O-acyltransferase 1 Oryctolagus cuniculus 265-269 3518714-2 1986 Alloxan-induced maternal diabetes (n = 5) produced mild fetal hyperinsulinemia (D) (plasma insulin concentrations = 59.80 +/- 8.10 microU/ml, control = 26.25 +/- 3.70; p less than 0.01), whereas systemic administration (IMI) of 1.0 U (n = 4) and 2.0 U (n = 4) of insulin to the fetus resulted in moderate (103.13 +/- 34.63 microU/ml) and severe (288.3 +/- 51 microU/ml) fetal hyperinsulinemia respectively. Alloxan 0-7 insulin Oryctolagus cuniculus 67-74 3527515-7 1986 In summary, after high doses of alloxan were given guinea pigs, serum insulin levels were reduced below control levels within 24 hr; this was associated with changes in body weight which paralleled changes in insulin levels more closely than did levels of blood or urine glucose; the volume density of immunostained B cells initially decreased and then returned to control levels by 72 hr following alloxan injection. Alloxan 32-39 insulin Cavia porcellus 70-77 3527515-7 1986 In summary, after high doses of alloxan were given guinea pigs, serum insulin levels were reduced below control levels within 24 hr; this was associated with changes in body weight which paralleled changes in insulin levels more closely than did levels of blood or urine glucose; the volume density of immunostained B cells initially decreased and then returned to control levels by 72 hr following alloxan injection. Alloxan 32-39 insulin Cavia porcellus 209-216 3530846-3 1986 Glucokinase partially purified from transplantable insulinomas or rat liver was inactivated by alloxan with a half-maximal effect at 2-4 microM alloxan. Alloxan 95-102 glucokinase Rattus norvegicus 0-11 3518714-2 1986 Alloxan-induced maternal diabetes (n = 5) produced mild fetal hyperinsulinemia (D) (plasma insulin concentrations = 59.80 +/- 8.10 microU/ml, control = 26.25 +/- 3.70; p less than 0.01), whereas systemic administration (IMI) of 1.0 U (n = 4) and 2.0 U (n = 4) of insulin to the fetus resulted in moderate (103.13 +/- 34.63 microU/ml) and severe (288.3 +/- 51 microU/ml) fetal hyperinsulinemia respectively. Alloxan 0-7 insulin Oryctolagus cuniculus 91-98 3962581-3 1986 Injection of alloxan resulted in a diabetic state characterized by increased hemoglobin glycosylation, blood and urine glucose and a significant depression of serum insulin levels. Alloxan 13-20 insulin Oryctolagus cuniculus 165-172 3158657-3 1985 Alloxan does not inhibit the uptake of Ca2+ but stimulates the release of Ca2+ from liver mitochondria, which is accompanied by oxidation and hydrolysis of pyridine nucleotides. Alloxan 0-7 carbonic anhydrase 2 Rattus norvegicus 74-77 4080455-1 1985 Diabetes induced by alloxan at day 6 of gestation in Wistar rats produced decreased fetal growth, delayed skeletal ossification, decreased fetal kidney beta-glucuronidase, and an increased frequency of fetal birth defects which correlated with the degree of diabetic control. Alloxan 20-27 glucuronidase, beta Rattus norvegicus 152-170 3158657-7 1985 It is concluded that alloxan can cause Ca2+ release from intact rat liver mitochondria. Alloxan 21-28 carbonic anhydrase 2 Rattus norvegicus 39-42 3158657-9 1985 Oxidation and hydrolysis of pyridine nucleotides, possibly in conjunction with oxidation of critical sulfhydryl groups, seem to be key events in the alloxan-induced Ca2+ release. Alloxan 149-156 carbonic anhydrase 2 Rattus norvegicus 165-168 6497140-7 1984 Sodium and chloride values were depressed in steers with alloxan-induced diabetes; these values remained significantly (P less than 0.05) lower than base-line values, even in steers given insulin. Alloxan 57-64 insulin Homo sapiens 188-195 3883362-1 1985 In insulin-deprived alloxan-induced diabetic dogs with severe hyperglycemia and marked hyperglucagonemia, glucagon was not suppressed by intravenous infusion of glucose at a progressively increasing rate up to 24 mg/kg of body weight per min. Alloxan 20-27 insulin Canis lupus familiaris 3-10 6240981-0 1984 Inhibition of glucokinase in hepatocytes by alloxan. Alloxan 44-51 glucokinase Rattus norvegicus 14-25 6240981-1 1984 The effect of alloxan on glucokinase in isolated rat hepatocytes was studied. Alloxan 14-21 glucokinase Rattus norvegicus 25-36 6240981-2 1984 Exposure of hepatocytes to alloxan (3 mM) at 30 degrees C for 5 min produced a marked inhibition (77%) of glucokinase activity and altered slightly the phosphofructokinase activity (32% inhibition). Alloxan 27-34 glucokinase Rattus norvegicus 106-117 6240981-7 1984 These results suggest that alloxan may exert its cytotoxic action through the inhibition of glucokinase activity not only in the liver but also in the pancreatic islets, since liver and islet glucokinases are known to be quite similar in various properties. Alloxan 27-34 glucokinase Rattus norvegicus 92-103 6510522-2 1984 The following approximative alloxan concentrations induced 50% inhibition of enzyme activity: 10(-6)M for aconitase, 10(-4)M for NAD-linked isocitrate dehydrogenase, glutamate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinyl-CoA synthetase and fumarase, and 10(-3)M for citrate synthase and NADP-linked isocitrate dehydrogenase. Alloxan 28-35 oxoglutarate (alpha-ketoglutarate) dehydrogenase (lipoamide) Mus musculus 191-224 6510522-2 1984 The following approximative alloxan concentrations induced 50% inhibition of enzyme activity: 10(-6)M for aconitase, 10(-4)M for NAD-linked isocitrate dehydrogenase, glutamate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinyl-CoA synthetase and fumarase, and 10(-3)M for citrate synthase and NADP-linked isocitrate dehydrogenase. Alloxan 28-35 fumarate hydratase 1 Mus musculus 254-262 6510522-2 1984 The following approximative alloxan concentrations induced 50% inhibition of enzyme activity: 10(-6)M for aconitase, 10(-4)M for NAD-linked isocitrate dehydrogenase, glutamate dehydrogenase, alpha-ketoglutarate dehydrogenase, succinyl-CoA synthetase and fumarase, and 10(-3)M for citrate synthase and NADP-linked isocitrate dehydrogenase. Alloxan 28-35 citrate synthase Mus musculus 280-296 6237009-3 1984 Exposure of islets to alloxan (2 mM) in vitro caused a marked inhibition of both glucose-stimulated proinsulin biosynthesis and insulin release, and this was not affected by the action of nicotinamide. Alloxan 22-29 insulin II Mus musculus 100-110 6237009-10 1984 The inability of nicotinamide to prevent the alloxan-induced impairment of proinsulin biosynthesis, insulin release, and oxygen uptake, together with the failure of nicotinamide to prevent the development of diabetes when given after alloxan, does not support a current hypothesis that the major cytotoxic effect of alloxan is primarily due to DNA damage. Alloxan 45-52 insulin II Mus musculus 75-85 6240183-6 1984 The activities of liver beta-glucuronidase, beta-N-acetyl glucosaminidase and cathepsin D were significantly increased in rats treated with streptozotocin and alloxan. Alloxan 159-166 glucuronidase, beta Rattus norvegicus 24-42 6351093-5 1983 Furthermore, insulin release from guinea pig but not rat cultures increased transiently at between 6 and 18 hr during the first day following exposure to all doses of alloxan. Alloxan 167-174 insulin Cavia porcellus 13-20 6240183-6 1984 The activities of liver beta-glucuronidase, beta-N-acetyl glucosaminidase and cathepsin D were significantly increased in rats treated with streptozotocin and alloxan. Alloxan 159-166 cathepsin D Rattus norvegicus 78-89 6363170-1 1984 The pretreatment of isolated islets of Langerhans with concanavalin A (Con A) completely blocks alloxan from suppressing the insulin release response to glucose. Alloxan 96-103 insulin Homo sapiens 125-132 6420098-3 1984 Three parameters of cell-mediated immunity, namely, (a) resistance to infection with Candida albicans, (b) in vivo release of migration inhibitory factor (MIF) into the circulation and (c) delayed hypersensitivity were markedly reduced when mice of such normally resistant high responder strains as C57B1/10SNJ and C57B1/KsJ became hyperglycaemic after treatment with alloxan. Alloxan 368-375 macrophage migration inhibitory factor (glycosylation-inhibiting factor) Mus musculus 155-158 6404681-1 1983 This study examined the effect of an aldose reductase inhibitor (Sorbinil, CP 45634, Pfizer, Sandwich, Kent, United Kingdom) on the metabolite profile of the lens during the first week after induction of diabetes with alloxan. Alloxan 218-225 aldo-keto reductase family 1 member B1 Rattus norvegicus 37-53 6309589-4 1983 Comparison of the alloxan- or streptozotocin-treated groups with control animals showed an increase in lysosomal enzymes (cathepsin B1, cathepsin D, beta-glucuronidase and beta-N-acetyl glucosaminidase in skin, liver and spleen) (p less than 0.05) but beta-N-acetyl glucosaminidase was unchanged in the spleen of streptozotocin-diabetic rats. Alloxan 18-25 cathepsin B Rattus norvegicus 122-134 6309589-4 1983 Comparison of the alloxan- or streptozotocin-treated groups with control animals showed an increase in lysosomal enzymes (cathepsin B1, cathepsin D, beta-glucuronidase and beta-N-acetyl glucosaminidase in skin, liver and spleen) (p less than 0.05) but beta-N-acetyl glucosaminidase was unchanged in the spleen of streptozotocin-diabetic rats. Alloxan 18-25 cathepsin D Rattus norvegicus 136-147 6309589-4 1983 Comparison of the alloxan- or streptozotocin-treated groups with control animals showed an increase in lysosomal enzymes (cathepsin B1, cathepsin D, beta-glucuronidase and beta-N-acetyl glucosaminidase in skin, liver and spleen) (p less than 0.05) but beta-N-acetyl glucosaminidase was unchanged in the spleen of streptozotocin-diabetic rats. Alloxan 18-25 glucuronidase, beta Rattus norvegicus 149-167 6341516-5 1983 A large surge of insulin release occurred immediately after alloxan administration, which was followed by a decrease in insulin concentrations to subnormal levels in those animals not treated with insulin. Alloxan 60-67 insulin Bos taurus 17-24 6341516-5 1983 A large surge of insulin release occurred immediately after alloxan administration, which was followed by a decrease in insulin concentrations to subnormal levels in those animals not treated with insulin. Alloxan 60-67 insulin Bos taurus 120-127 6341516-5 1983 A large surge of insulin release occurred immediately after alloxan administration, which was followed by a decrease in insulin concentrations to subnormal levels in those animals not treated with insulin. Alloxan 60-67 insulin Bos taurus 120-127 6753106-2 1982 Compared with the controls, the methyl-prednisolone-treated rats had increased fasting levels of serum insulin, blood glucose, and plasma GIP; the alloxan-treated rats had decreased fasting levels of insulin and increased levels of fasting blood glucose and plasma GIP. Alloxan 147-154 gastric inhibitory polypeptide Rattus norvegicus 265-268 7042326-1 1982 Noncarbohydrate nutrients such as 2-ketoisocaproate, L-glutamine, L-leucine and its nonmetabolized analog, beta-2-aminobicyclo[2,2,1]heptane-2-carboxylic acid (BCH) were able to mimic, to a limited extent, the protective action of D-glucose against the inhibitory effect of alloxan on glucose-stimulated insulin release. Alloxan 274-281 potassium calcium-activated channel subfamily M regulatory beta subunit 2 Homo sapiens 107-113 7159455-2 1982 At concentrations of 3.5 mM and above, alloxan caused an increase in lactate dehydrogenase (LDH), glutamate-pyruvate transaminase (GPT) and intracellular potassium (K+) leakage, all of which are indices of plasma membrane damage, and decreased the intracellular reduced glutathione content (GSH) of the cells. Alloxan 39-46 glutamic--pyruvic transaminase Rattus norvegicus 98-129 7159455-2 1982 At concentrations of 3.5 mM and above, alloxan caused an increase in lactate dehydrogenase (LDH), glutamate-pyruvate transaminase (GPT) and intracellular potassium (K+) leakage, all of which are indices of plasma membrane damage, and decreased the intracellular reduced glutathione content (GSH) of the cells. Alloxan 39-46 glutamic--pyruvic transaminase Rattus norvegicus 131-134 6753106-5 1982 The elevated levels of fasting plasma GIP in the diabetic rats treated with methylprednisolone or alloxan may be due to factors regulating the secretion of GIP rather than to changes in the duodenal content of GIP. Alloxan 98-105 gastric inhibitory polypeptide Rattus norvegicus 38-41 6753106-5 1982 The elevated levels of fasting plasma GIP in the diabetic rats treated with methylprednisolone or alloxan may be due to factors regulating the secretion of GIP rather than to changes in the duodenal content of GIP. Alloxan 98-105 gastric inhibitory polypeptide Rattus norvegicus 156-159 6753106-5 1982 The elevated levels of fasting plasma GIP in the diabetic rats treated with methylprednisolone or alloxan may be due to factors regulating the secretion of GIP rather than to changes in the duodenal content of GIP. Alloxan 98-105 gastric inhibitory polypeptide Rattus norvegicus 156-159 6272129-0 1981 Streptozotocin and alloxan induce DNA strand breaks and poly(ADP-ribose) synthetase in pancreatic islets. Alloxan 19-26 poly(ADP-ribose) polymerase 1 Homo sapiens 56-83 6753105-4 1982 Similarly, treatment with alloxan was followed by decreased fasting levels of serum insulin, by increased fasting levels of blood glucose and plasma GIP, and by an increased GIP release in response to duodenal glucose and amino acids. Alloxan 26-33 gastric inhibitory polypeptide Rattus norvegicus 149-152 6753105-4 1982 Similarly, treatment with alloxan was followed by decreased fasting levels of serum insulin, by increased fasting levels of blood glucose and plasma GIP, and by an increased GIP release in response to duodenal glucose and amino acids. Alloxan 26-33 gastric inhibitory polypeptide Rattus norvegicus 174-177 6753105-5 1982 The augmented GIP release in response to duodenal instillation of glucose and amino acids both in methylprednisolone-treated rats and in alloxan-treated rats may be explained by an increased absorption of these nutrients owing to an increased Na+ K+ ATPase activity in the intestinal mucosa of corticosteroid- and alloxan-treated rats. Alloxan 137-144 gastric inhibitory polypeptide Rattus norvegicus 14-17 7041886-1 1981 Exogenous superoxide dismutase, catalase and scavengers of the hydroxyl radical protect pancreatic-islet cells against the toxic actions of alloxan in vitro [Grankvist et al. Alloxan 140-147 catalase Mus musculus 32-40 6933551-5 1980 Thus, alloxan showed an apparent Km of 70 microM in the presence of 3.4 microM E. coli thioredoxin, 0.2 microM thioredoxin reductase, and 0.4 mM NADPH. Alloxan 6-13 thioredoxin Bos taurus 87-98 7024025-11 1981 Rapid reduction of alloxan by thioredoxin in the presence of molecular oxygen and NADPH leads to strong chemiluminescence from luminol indicative of an intense radical protection. Alloxan 19-26 thioredoxin 1 Mus musculus 30-41 7027617-2 1981 The hypoglycemic effect of insulin, included into this complex as compared with free insulin, was increased more than 2-fold when it was tested in animals with stable alloxane diabetes. Alloxan 167-175 insulin Homo sapiens 27-34 6933551-5 1980 Thus, alloxan showed an apparent Km of 70 microM in the presence of 3.4 microM E. coli thioredoxin, 0.2 microM thioredoxin reductase, and 0.4 mM NADPH. Alloxan 6-13 thioredoxin Bos taurus 111-122 232388-0 1979 Kinetic studies of the blood serum xanthine dehydrogenase inhibition by alloxan (2,4,5,6-tetraoxypyrimidine 5,6-dioxyuracil). Alloxan 72-79 xanthine dehydrogenase Homo sapiens 35-57 6248397-3 1980 A single injection of alloxan (65 mg/kg) led, after 6 weeks, to a 40% decrease of testicular LH- and prolactin-receptor levels. Alloxan 22-29 luteinizing hormone/choriogonadotropin receptor Rattus norvegicus 93-119 6444670-1 1980 Dopamine-beta-hydroxylase (DBH) activity is increased at least 5-fold in the serum of rats with experimental diabetes produced by Streptozotocin or Alloxan. Alloxan 148-155 dopamine beta-hydroxylase Rattus norvegicus 0-25 6444670-1 1980 Dopamine-beta-hydroxylase (DBH) activity is increased at least 5-fold in the serum of rats with experimental diabetes produced by Streptozotocin or Alloxan. Alloxan 148-155 dopamine beta-hydroxylase Rattus norvegicus 27-30 7449751-2 1980 Ornithine decarboxylase (L-ornithine carboxylase; EC 4.1.1.17) (ODC) activity falls to approximately 30% of the control value in diabetic rat kidney 4 weeks after induction of diabetes with alloxan. Alloxan 190-197 ornithine decarboxylase 1 Rattus norvegicus 0-23 7449751-2 1980 Ornithine decarboxylase (L-ornithine carboxylase; EC 4.1.1.17) (ODC) activity falls to approximately 30% of the control value in diabetic rat kidney 4 weeks after induction of diabetes with alloxan. Alloxan 190-197 ornithine decarboxylase 1 Rattus norvegicus 64-67 40548-0 1979 Superoxide dismutase, catalase and scavengers of hydroxyl radical protect against the toxic action of alloxan on pancreatic islet cells in vitro. Alloxan 102-109 catalase Mus musculus 22-30 40548-6 1979 Superoxide dismutase, catalase, dimethyl sulphoxide, benzoate, and mannitol counteracted the effects of alloxan in both cytotoxicity assays. Alloxan 104-111 catalase Mus musculus 22-30 294757-1 1979 Xanthine dehydrogenase activity was determined in blood serum of rats in which diabetes had been induced by alloxan administration. Alloxan 108-115 xanthine dehydrogenase Rattus norvegicus 0-22 465017-0 1979 Effect of alloxan-induced chronic diabetes on lipid composition and lipoprotein lipase activity of rat lung. Alloxan 10-17 lipoprotein lipase Rattus norvegicus 68-86 321126-0 1977 Effect of alloxan on rat pancreatic polypeptide (PP) cells. Alloxan 10-17 pancreatic polypeptide Rattus norvegicus 25-47 711132-5 1978 SPX animals contrastingly showed significantly less response to alloxan than did animals of the other two groups. Alloxan 64-71 spexin hormone Rattus norvegicus 0-3 676768-2 1978 After treatment of the diabetic goats with insulin for 4--5 days--the last 24 h intravenously--lactose secretion returned to the control values before alloxan administration provided that normoglycemia developed. Alloxan 151-158 insulin Capra hircus 43-50 664469-2 1978 In diabetes the activity of these enzymes in rat kidney, as distinct from liver tissue, was not decreased but it was elevated and within 72 hrs after administration of alloxan the activity of glucose-6-phosphate dehydrogenase was increased 2-fold and the activity of 6-phosphogluconate dehydrogenase was increased by 30% above the normal level. Alloxan 168-175 glucose-6-phosphate dehydrogenase Rattus norvegicus 192-225 331860-4 1977 Starved mice injected with DHCC or PTH 10 minutes before alloxan administration exhibited a pronounced second hyperglycemia of long duration, and extensive, selective B-cell necrosis. Alloxan 57-64 parathyroid hormone Mus musculus 35-38 186585-8 1976 Dithiobisnitrobenzoic acid and alloxan, inhibitors of adenylate cyclase, impaired neuromuscular transmission and prevented the effects of NaF, but they did not change the responses to dibutyryl cAMP. Alloxan 31-38 C-X-C motif chemokine ligand 8 Homo sapiens 138-141 608581-2 1977 The activities of these enzymes were increased significantly in the alloxan-treated rats kept in LD (light: darkness) cycles of 10:14 h. Continuous light exposure to diabetic animals appeared to decrease delta5-3beta-HSD and g-6-PDH in comparison to the diabetic rats kept in 10 h illumination. Alloxan 68-75 glucose-6-phosphate dehydrogenase Rattus norvegicus 225-232 138586-1 1977 Duodenal calcium absorption and calcium binding protein (CaBP) are depressed in uncontrolled experimental (alloxan and streptozotocin) diabeties in the rat. Alloxan 107-114 S100 calcium binding protein G Rattus norvegicus 9-55 138586-1 1977 Duodenal calcium absorption and calcium binding protein (CaBP) are depressed in uncontrolled experimental (alloxan and streptozotocin) diabeties in the rat. Alloxan 107-114 S100 calcium binding protein G Rattus norvegicus 57-61 4278073-0 1974 Effect of insulin infusions on the glucose kinetics in alloxan-steptozotocin diabetic dogs. Alloxan 55-62 insulin Canis lupus familiaris 10-17 128691-0 1975 Inhibition of insulin biosynthesis by alloxan, streptozotocin, and N-nitrosomethylurea. Alloxan 38-45 insulin Homo sapiens 14-21 164533-1 1975 Studies were performed to examine the effects of alloxan- or streptozotocin-induced diabetes on carbon tetrachloride (CCl4) liver injury. Alloxan 49-56 C-C motif chemokine ligand 4 Rattus norvegicus 118-122 164533-12 1975 Insulin treatment of rats given alloxan (80 mg/kg) markedly protected against CCl4-induced hepatotoxicity. Alloxan 32-39 C-C motif chemokine ligand 4 Rattus norvegicus 78-82 5599806-0 1967 [Glucose-6-phosphate dehydrogenase and 6-phosphogluconate dehydrogenase activity in epididymal adipose tissue from normal rats, fasted rats and rats with alloxan-induced diabetes]. Alloxan 154-161 glucose-6-phosphate dehydrogenase Rattus norvegicus 1-34 4588113-0 1973 Acute effects of alloxan on the metabolism and insulin secretion of the pancreatic B-cell. Alloxan 17-24 insulin Homo sapiens 47-54 24683634-1 1972 The extremely high levels of glucagon recently observed in dogs with severe alloxan-induced diabetes decline promptly and precipitously to normal as soon as exogenous insulin is infused. Alloxan 76-83 insulin Canis lupus familiaris 167-174 4311516-0 1968 Nature of the increase in liver microsomal glucose-6-phosphatase activity during the early stages of alloxan-induced diabetes. Alloxan 101-108 glucose-6-phosphatase catalytic subunit 1 Homo sapiens 43-64 4590866-0 1973 [Effect of alloxan on the content of insulin and zinc in the pancreatic islets in experimental animals]. Alloxan 11-18 insulin Homo sapiens 37-44 4248447-0 1970 C14-proline metabolism in alloxan treated rats. Alloxan 26-33 anti-Mullerian hormone receptor type 2 Rattus norvegicus 0-3 13792172-2 1959 after the maintenance dose of insulin (28.1%+/-2.9 s.e) in rabbits made severely diabetic with alloxan was significantly different from the change in the blood sugar with the same drug given 72 hr. Alloxan 95-102 insulin Oryctolagus cuniculus 30-37 13800358-0 1959 Inhibition of liver hexokinase by dehydroascorbic acid and alloxan. Alloxan 59-66 hexokinase 1 Homo sapiens 20-30 13886629-0 1962 Effect of alloxan on the insulin content of micro-dissected mammalian pancreatic islets. Alloxan 10-17 insulin Homo sapiens 25-32 13463225-0 1957 The preventive effect of glucagon (HGF) against the diabetogenic action of alloxan and its antagonism by insulin. Alloxan 75-82 hepatocyte growth factor Homo sapiens 35-38 13544948-0 1958 [Studies on the reduction of alloxan and the oxidation of dialuric acid in the presence of alcohol dehydrogenase]. Alloxan 29-36 aldo-keto reductase family 1 member A1 Homo sapiens 91-112 20286883-0 1946 Effect of alloxan on insulin. Alloxan 10-17 insulin Homo sapiens 21-28 14790813-0 1950 Oxidation of uric acid and of alloxan by cytochrome c. Alloxan 30-37 cytochrome c, somatic Homo sapiens 41-53 13201714-5 1954 It was therefore concluded that the inhibitory phenomenon was not due to the injection of alloxan per se but that it was associated with one or more factors that characterize the alloxan diabetic state in the rabbit and that are reversible by insulin therapy. Alloxan 179-186 insulin Oryctolagus cuniculus 243-250 33470760-10 2021 It is worth noting that the expression of HSP70 and the protein O-GlcNAc modification levels in the Gln-treated group were significantly elevated than the levels in the sepsis group (P < 0.05), and reversed by pretreatment with the HSP and O-GlcNAc inhibitors quercetion and alloxan. Alloxan 275-282 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 42-47 14839139-0 1950 [Action of the alloxan-dialuric acid system on the activity of cholinesterase in vitro and in vivo]. Alloxan 15-22 butyrylcholinesterase Homo sapiens 63-77 34015900-8 2021 Interestingly, compared with normal control levels, S1P/EDG-5 was increased in both alloxan and glucose treated pancreatic beta cell than normal control. Alloxan 84-91 sphingosine-1-phosphate receptor 2 Mus musculus 56-61 34015900-9 2021 MAPK-ERK inhibition strongly decreased the expression of insulin and S1P in glucose- or alloxan-treated RIN-5F cells. Alloxan 88-95 mitogen-activated protein kinase 1 Mus musculus 5-8 32911700-1 2020 In our study, we aimed to evaluate the effects of Moringa oleifera leaves extract on rat paraoxonase 1 (rPON1) and catalase (rCAT) activities in alloxan-induced diabetic rats. Alloxan 145-152 paraoxonase 1 Rattus norvegicus 89-102 33039567-6 2021 Results of the mitotic-index observed from the PG-pre-treated-alloxan-administered (PG+ALX) mice group revealed a significant reduction in chromosomal-anomaly, DNA-damage, and an upregulation of the p53 and PARP protein expression when compared to the ALX-treated mice group. Alloxan 62-69 transformation related protein 53, pseudogene Mus musculus 199-202 33039567-6 2021 Results of the mitotic-index observed from the PG-pre-treated-alloxan-administered (PG+ALX) mice group revealed a significant reduction in chromosomal-anomaly, DNA-damage, and an upregulation of the p53 and PARP protein expression when compared to the ALX-treated mice group. Alloxan 62-69 poly (ADP-ribose) polymerase family, member 1 Mus musculus 207-211 32534114-22 2020 RESULTS: Alloxan treatment increased the levels of FBG, total cholesterol, LDL-cholesterol, VLDL-cholesterol, urine ketone and cardiac function indices and reduced the levels of globulin, albumin, HDL-cholesterol, globulin, liver glycogen, hexokinase and glucose-6-phosphate dehydrogenase activities. Alloxan 9-16 glucose-6-phosphate dehydrogenase Rattus norvegicus 255-288 32440769-0 2020 Pyrrolidine dithiocarbamate reduces alloxan-induced kidney damage by decreasing nox4, inducible nitric oxide synthase, and metalloproteinase-2. Alloxan 36-43 NADPH oxidase 4 Rattus norvegicus 80-84 32440769-0 2020 Pyrrolidine dithiocarbamate reduces alloxan-induced kidney damage by decreasing nox4, inducible nitric oxide synthase, and metalloproteinase-2. Alloxan 36-43 nitric oxide synthase 2 Rattus norvegicus 86-117 33359794-6 2021 More importantly, the protective effects of G-Rh4 on alloxan-induced upregulation of Nrf2 target gene and insulin secretion were abolished by Nrf2 knockdown. Alloxan 53-60 nuclear factor, erythroid derived 2, like 2 Mus musculus 85-89 33359794-6 2021 More importantly, the protective effects of G-Rh4 on alloxan-induced upregulation of Nrf2 target gene and insulin secretion were abolished by Nrf2 knockdown. Alloxan 53-60 nuclear factor, erythroid derived 2, like 2 Mus musculus 142-146 32979266-4 2020 Insulin-dependent diabetes was induced by alloxan in female rabbits. Alloxan 42-49 insulin Oryctolagus cuniculus 0-7 32911700-1 2020 In our study, we aimed to evaluate the effects of Moringa oleifera leaves extract on rat paraoxonase 1 (rPON1) and catalase (rCAT) activities in alloxan-induced diabetic rats. Alloxan 145-152 paraoxonase 1 Rattus norvegicus 104-109 32908936-0 2020 Peroxiredoxin 6 Attenuates Alloxan-Induced Type 1 Diabetes Mellitus in Mice and Cytokine-Induced Cytotoxicity in RIN-m5F Beta Cells. Alloxan 27-34 peroxiredoxin 6 Mus musculus 0-15 32954201-1 2020 Alloxan (AL) is a toxic glucose analogue that acts as a potent diabetogenic inducer by selectively destroying the insulin-producing beta-cells of the pancreas. Alloxan 0-7 insulin Homo sapiens 114-121 32954201-1 2020 Alloxan (AL) is a toxic glucose analogue that acts as a potent diabetogenic inducer by selectively destroying the insulin-producing beta-cells of the pancreas. Alloxan 9-11 insulin Homo sapiens 114-121 32908936-5 2020 It was shown that PRDX6 prevented hyperglycemia, lowered the mortality rate, restored the plasma cytokine profile, reversed the splenic cell apoptosis, and reduced the beta cell destruction in Langerhans islets in mice with a severe form of alloxan-induced diabetes. Alloxan 241-248 peroxiredoxin 6 Mus musculus 18-23 32422500-0 2020 Polysaccharides from Opuntia milpa alta alleviate alloxan-induced INS-1 cells apoptosis via reducing oxidative stress and upregulating Nrf2 expression. Alloxan 50-57 NFE2 like bZIP transcription factor 2 Rattus norvegicus 135-139 32454931-0 2020 Mulberry Fruit Prevents Diabetes and Diabetic Dementia by Regulation of Blood Glucose through Upregulation of Antioxidative Activities and CREB/BDNF Pathway in Alloxan-Induced Diabetic Mice. Alloxan 160-167 cAMP responsive element binding protein 1 Mus musculus 139-143 32454931-0 2020 Mulberry Fruit Prevents Diabetes and Diabetic Dementia by Regulation of Blood Glucose through Upregulation of Antioxidative Activities and CREB/BDNF Pathway in Alloxan-Induced Diabetic Mice. Alloxan 160-167 brain derived neurotrophic factor Mus musculus 144-148 32422500-7 2020 We found that MAPs could attenuate alloxan-induced apoptosis by increasing the expression of Bcl-2 and decreasing the expression of Bax and the activities of caspase-3 and caspase-9. Alloxan 35-42 BCL2, apoptosis regulator Rattus norvegicus 93-98 32422500-7 2020 We found that MAPs could attenuate alloxan-induced apoptosis by increasing the expression of Bcl-2 and decreasing the expression of Bax and the activities of caspase-3 and caspase-9. Alloxan 35-42 BCL2 associated X, apoptosis regulator Rattus norvegicus 132-135 32422500-7 2020 We found that MAPs could attenuate alloxan-induced apoptosis by increasing the expression of Bcl-2 and decreasing the expression of Bax and the activities of caspase-3 and caspase-9. Alloxan 35-42 caspase 3 Rattus norvegicus 158-167 32422500-7 2020 We found that MAPs could attenuate alloxan-induced apoptosis by increasing the expression of Bcl-2 and decreasing the expression of Bax and the activities of caspase-3 and caspase-9. Alloxan 35-42 caspase 9 Rattus norvegicus 172-181 32422500-9 2020 These findings indicated that MAPs could alleviate alloxan-induced beta-cell apoptosis by reducing oxidative stress and upregulating Nrf2 expression. Alloxan 51-58 NFE2 like bZIP transcription factor 2 Rattus norvegicus 133-137 31637753-0 2019 Tlr1-13, Nod1/2 and antimicrobial gene expression in the epididymis and testis of rats with alloxan-induced diabetes. Alloxan 92-99 toll-like receptor 1 Rattus norvegicus 0-4 33728270-2 2021 To this end, the antidiabetic activity of aqueous stem-bark extract of A. polycarpa (APE) in alloxan-induced diabetic ICR mice was investigated. Alloxan 93-100 apurinic/apyrimidinic endonuclease 1 Mus musculus 85-88 32019238-3 2020 Insulin-dependent diabetes was induced by alloxan treatment in female rabbits 10 days before mating. Alloxan 42-49 insulin Oryctolagus cuniculus 0-7 31823816-6 2019 RESULTS: BDNF, LXA4 and AA, EPA and DHA protected (P < 0.001 and P < 0.01 respectively) against AL/STZ/DB/BP-induced toxicity to RIN5F cells in vitro. Alloxan 102-104 brain derived neurotrophic factor Mus musculus 9-13 31823816-7 2019 AL/ STZ/DB/BP inhibited BDNF and LXA4 production by RIN5F cells and were restored to normal by AA, EPA and DHA. Alloxan 0-2 brain derived neurotrophic factor Mus musculus 24-28 31823816-8 2019 Sub-optimal doses of BDNF, LXA4, AA and EPA when used in combination protected against cytotoxic action of AL/STZ/DB/BP on RIN5F cells in vitro by restoring LXA4/BDNF levels and altered antioxidant/lipid peroxides/NO levels (P < 0.01) to normal. Alloxan 107-109 brain derived neurotrophic factor Mus musculus 21-25 31823816-8 2019 Sub-optimal doses of BDNF, LXA4, AA and EPA when used in combination protected against cytotoxic action of AL/STZ/DB/BP on RIN5F cells in vitro by restoring LXA4/BDNF levels and altered antioxidant/lipid peroxides/NO levels (P < 0.01) to normal. Alloxan 107-109 brain derived neurotrophic factor Mus musculus 162-166 31823816-10 2019 DISCUSSION: AL/STZ/DB/BP-induced cytotoxicity to RIN5F cells in vitro can be prevented by BDNF, LXA4 and AA. Alloxan 12-14 brain derived neurotrophic factor Mus musculus 90-94 31823816-11 2019 AL/STZ/DB/BP are cytotoxic, possibly, by suppressing the production of LXA4 and BDNF in RIN5F cells. Alloxan 0-2 brain derived neurotrophic factor Mus musculus 80-84 32256963-0 2020 Effect of Caesalpinia bonduc Polyphenol Extract on Alloxan-Induced Diabetic Rats in Attenuating Hyperglycemia by Upregulating Insulin Secretion and Inhibiting JNK Signaling Pathway. Alloxan 51-58 mitogen-activated protein kinase 8 Rattus norvegicus 159-162 31738423-6 2020 Fluorescence microscopy revealed that alloxan interferes with the motility of cellular organelles (peroxisomes, endosomes and the endoplasmic reticulum) and the pathogen-triggered redistribution of the PEN1/SYP121 t-SNARE protein. Alloxan 38-45 pentacyclic triterpene synthase 1 Arabidopsis thaliana 202-206 31738423-6 2020 Fluorescence microscopy revealed that alloxan interferes with the motility of cellular organelles (peroxisomes, endosomes and the endoplasmic reticulum) and the pathogen-triggered redistribution of the PEN1/SYP121 t-SNARE protein. Alloxan 38-45 syntaxin of plants 121 Arabidopsis thaliana 207-213 31378691-8 2020 CONCLUSIONS: Instant tea and matcha supplementation had beneficial effects on regulation of blood glucose and gut microbiota, reversing the changes in microbiota caused by alloxan injection. Alloxan 172-179 solute carrier family 7 (cationic amino acid transporter, y+ system), member 2 Mus musculus 21-24 32155637-4 2020 OBJECTIVES: We investigated herein the expression and role of semaphorin-3A in the migratory responses of thymocytes from alloxan-induced diabetic mice. Alloxan 122-129 sema domain, immunoglobulin domain (Ig), short basic domain, secreted, (semaphorin) 3A Mus musculus 62-75 30465800-5 2019 Alloxan-induced diabetic wild- type mice (diabetic) developed wounds that were difficult to heal, differently from CCR4-/- diabetic mice (CCR4-/- diabetic), and also from anti-CCL17/22 or anti-CD25-injected diabetic mice that presented with accelerated wound healing and fewer regulatory T cells in the wound bed. Alloxan 0-7 chemokine (C-C motif) receptor 4 Mus musculus 115-119 31827688-3 2019 Here, we report that increased O-GlcNAcylation by the suppression of OGA activity using thiamet-G and OGA siRNA did not affect autophagy, whereas decreased O-GlcNAcylation caused by OGT inhibition by alloxan and OGT siRNA increased autophagy. Alloxan 200-207 O-linked N-acetylglucosamine (GlcNAc) transferase (UDP-N-acetylglucosamine:polypeptide-N-acetylglucosaminyl transferase) Mus musculus 182-185 31827688-6 2019 Additionally, decreasing O-GlcNAcylation by treatment with alloxan, OGT siRNA, and OGA overexpression significantly decreased the level of autophagy substrate SQSTM1/p62, indicating that autophagic degradation was activated. Alloxan 59-66 sequestosome 1 Mus musculus 159-165 31827688-6 2019 Additionally, decreasing O-GlcNAcylation by treatment with alloxan, OGT siRNA, and OGA overexpression significantly decreased the level of autophagy substrate SQSTM1/p62, indicating that autophagic degradation was activated. Alloxan 59-66 sequestosome 1 Mus musculus 166-169 30465800-5 2019 Alloxan-induced diabetic wild- type mice (diabetic) developed wounds that were difficult to heal, differently from CCR4-/- diabetic mice (CCR4-/- diabetic), and also from anti-CCL17/22 or anti-CD25-injected diabetic mice that presented with accelerated wound healing and fewer regulatory T cells in the wound bed. Alloxan 0-7 interleukin 2 receptor, alpha chain Mus musculus 193-197 30465800-5 2019 Alloxan-induced diabetic wild- type mice (diabetic) developed wounds that were difficult to heal, differently from CCR4-/- diabetic mice (CCR4-/- diabetic), and also from anti-CCL17/22 or anti-CD25-injected diabetic mice that presented with accelerated wound healing and fewer regulatory T cells in the wound bed. Alloxan 0-7 chemokine (C-C motif) ligand 17 Mus musculus 176-184 30841444-0 2019 Antidiabetic role of a novel protein from garlic via NO in expression of Glut-4/insulin in liver of alloxan induced diabetic mice. Alloxan 100-107 solute carrier family 2 (facilitated glucose transporter), member 4 Mus musculus 73-79 30894246-9 2019 However, compared with adult pigs, juvenile swine exhibited greater insulin response recovery (complete or partial restoration of peripheral insulin levels to reference values) at 6 mo after alloxan administration. Alloxan 191-198 insulin Sus scrofa 68-75 30894246-10 2019 Overall, these results indicate that youth can confer some protection against the diabetogenic effects of alloxan in swine, potentially due in part to the greater insulin response recovery of young pigs. Alloxan 106-113 insulin Sus scrofa 163-170 30181664-4 2018 METHODS: The decrease of O-GlcNAcylation by alloxan, an OGT inhibitor, and increase by NAG-thiazolines (NAG-Ae), an O-GlcNAcase inhibitor were tested to investigate the effects of O-GlcNAc alteration on AD like neurodegeneration in SK-N-SH cells. Alloxan 44-51 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 56-59 30532634-13 2018 According to our findings we confirmed that zingerone restrained the alloxan induced oxidative stress by increasing the activity of reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX) and reducing the peroxidative damage. Alloxan 69-76 catalase Homo sapiens 187-195 30532634-13 2018 According to our findings we confirmed that zingerone restrained the alloxan induced oxidative stress by increasing the activity of reduced glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX) and reducing the peroxidative damage. Alloxan 69-76 catalase Homo sapiens 197-200 29421520-3 2018 hMSC-TERT supported survival of cocultured INS-1E beta-cells during cellular stress by alloxan (ALX) and streptozotocin (STZ), but not in response to IL-1beta. Alloxan 87-94 telomerase reverse transcriptase Rattus norvegicus 5-9 29421520-3 2018 hMSC-TERT supported survival of cocultured INS-1E beta-cells during cellular stress by alloxan (ALX) and streptozotocin (STZ), but not in response to IL-1beta. Alloxan 87-94 insulin 1 Rattus norvegicus 43-48 29421520-3 2018 hMSC-TERT supported survival of cocultured INS-1E beta-cells during cellular stress by alloxan (ALX) and streptozotocin (STZ), but not in response to IL-1beta. Alloxan 96-99 telomerase reverse transcriptase Rattus norvegicus 5-9 29421520-3 2018 hMSC-TERT supported survival of cocultured INS-1E beta-cells during cellular stress by alloxan (ALX) and streptozotocin (STZ), but not in response to IL-1beta. Alloxan 96-99 insulin 1 Rattus norvegicus 43-48 30016155-5 2018 Furthermore, NLRP3 and ASC-deficient mice were protected against oxidative stress-induced diabetes caused by repetitive low-dose alloxan administration, and this was associated with reduced beta-cell death and reduced macrophage infiltration. Alloxan 129-136 NLR family, pyrin domain containing 3 Mus musculus 13-18 30181664-4 2018 METHODS: The decrease of O-GlcNAcylation by alloxan, an OGT inhibitor, and increase by NAG-thiazolines (NAG-Ae), an O-GlcNAcase inhibitor were tested to investigate the effects of O-GlcNAc alteration on AD like neurodegeneration in SK-N-SH cells. Alloxan 44-51 O-linked N-acetylglucosamine (GlcNAc) transferase Homo sapiens 25-33 29784660-5 2018 beta-Cell-specific loss of IL-6 signaling in vivo renders mice more susceptible to oxidative damage and cell death through the selective beta-cell toxins streptozotocin and alloxan. Alloxan 173-180 interleukin 6 Homo sapiens 27-31 29929439-7 2018 In contrast, cyclin-B1 and cyclin-dependent kinase 1, mediators of G2 arrest, were remarkably changed in ALX-treated rats. Alloxan 105-108 cyclin B1 Rattus norvegicus 13-22 29929439-7 2018 In contrast, cyclin-B1 and cyclin-dependent kinase 1, mediators of G2 arrest, were remarkably changed in ALX-treated rats. Alloxan 105-108 cyclin-dependent kinase 1 Rattus norvegicus 27-52 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 90-93 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 10-38 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 90-93 heat shock protein family A (Hsp70) member 1A Rattus norvegicus 41-47 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 90-93 heat shock protein 90 alpha family class B member 1 Rattus norvegicus 50-58 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 90-93 heat shock protein family H (Hsp110) member 1 Rattus norvegicus 64-69 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 124-127 heat shock protein family A (Hsp70) member 1B Rattus norvegicus 10-38 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 124-127 heat shock protein family A (Hsp70) member 1A Rattus norvegicus 41-47 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 124-127 heat shock protein 90 alpha family class B member 1 Rattus norvegicus 50-58 29929439-9 2018 Moreover, heat-shock 70 kDA protein 1A ( Hspa1a), Hsp90ab1, and Hsph1 were upregulated in ALX-treated rats, suggesting that ALX treatment injures beta cells via endoplasmic reticulum stress. Alloxan 124-127 heat shock protein family H (Hsp110) member 1 Rattus norvegicus 64-69