PMID-sentid Pub_year Sent_text comp_official_name comp_offset protein_name organism prot_offset 16132793-1 2005 OBJECTIVE: The aim of this study was to investigate the role of alcohol dehydrogenase type 3 (ADH3), glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) polymorphisms in modifying hepatocellular carcinoma (HCC) risk according to alcohol intake. Alcohols 64-71 glutathione S-transferase theta 1 Homo sapiens 146-151 16308270-4 2005 Because the toxic effect of various chemicals can be modified by metabolic traits, the study also investigated the influence of the glutathione S-transferases (GSTM1 and GSTT1) on the toxic effect of DMF. Dimethylformamide 200-203 glutathione S-transferase theta 1 Homo sapiens 170-175 16308270-11 2005 Compared with the low DMF group with GSTT1-positive genotype workers, the odds ratio (adjusted for HBV status) of abnormal liver function test was 12.38, 95% CI=(1.04-146.9) for the high DMF group with GSTT1 null genotype workers. Dimethylformamide 22-25 glutathione S-transferase theta 1 Homo sapiens 37-42 16308270-11 2005 Compared with the low DMF group with GSTT1-positive genotype workers, the odds ratio (adjusted for HBV status) of abnormal liver function test was 12.38, 95% CI=(1.04-146.9) for the high DMF group with GSTT1 null genotype workers. Dimethylformamide 187-190 glutathione S-transferase theta 1 Homo sapiens 37-42 16308270-11 2005 Compared with the low DMF group with GSTT1-positive genotype workers, the odds ratio (adjusted for HBV status) of abnormal liver function test was 12.38, 95% CI=(1.04-146.9) for the high DMF group with GSTT1 null genotype workers. Dimethylformamide 187-190 glutathione S-transferase theta 1 Homo sapiens 202-207 16257343-2 2005 We therefore studied the role of cruciferous vegetables in lung cancer after stratifying by GSTM1 and GSTT1 status, two genes implicated in the elimination of isothiocyanates, the likely chemopreventative compound. Isothiocyanates 159-174 glutathione S-transferase theta 1 Homo sapiens 102-107 16173971-2 2005 Cellular sensitivity to 1,2:3,4 diepoxybutane was significantly increased in GSTT1 deleted compared with GSTT1 positive cases (median chromosomal breaks 11.1 vs. 8.3, P < 0.01) but there was no effect on clinical manifestations of FA. diepoxybutane 24-45 glutathione S-transferase theta 1 Homo sapiens 77-82 16173971-2 2005 Cellular sensitivity to 1,2:3,4 diepoxybutane was significantly increased in GSTT1 deleted compared with GSTT1 positive cases (median chromosomal breaks 11.1 vs. 8.3, P < 0.01) but there was no effect on clinical manifestations of FA. diepoxybutane 24-45 glutathione S-transferase theta 1 Homo sapiens 105-110 16133571-0 2005 The gut fermentation product butyrate, a chemopreventive agent, suppresses glutathione S-transferase theta (hGSTT1) and cell growth more in human colon adenoma (LT97) than tumor (HT29) cells. Butyrates 29-37 glutathione S-transferase theta 1 Homo sapiens 108-114 16133571-5 2005 Stability of GST-theta (hGSTT1) mRNA was assessed in HT29 cells after inhibition of transcription with actinomycin D. Dactinomycin 103-116 glutathione S-transferase theta 1 Homo sapiens 24-30 16133571-7 2005 Butyrate did not induce GSTs, but instead reduced hGSTT1 in LT97 and HT29. Butyrates 0-8 glutathione S-transferase theta 1 Homo sapiens 50-56 16133571-8 2005 CONCLUSIONS: Butyrate has suppressing-agent activities in human colon cells by inhibiting two survival factors, namely hGSTT1 and cell growth, with LT97 more sensitive than HT29. Butyrates 13-21 glutathione S-transferase theta 1 Homo sapiens 119-125 16536303-1 2005 OBJECTIVE: To study the association between susceptibility to aflatoxin B1 (AFB1)-related hepatocellular carcinoma(HCC) and the null genotypes of detoxication gene gstM1 and gstT1. Aflatoxin B1 62-74 glutathione S-transferase theta 1 Homo sapiens 174-179 16308270-12 2005 This study indicates that abnormal liver function and chronic liver disease are associated with DMF exposure, and there are more than multiplicative interaction effects on abnormal liver function tests between the DMF exposure and the GSTT1 genotype. Dimethylformamide 214-217 glutathione S-transferase theta 1 Homo sapiens 235-240 15790493-0 2005 Genotoxicity and metabolism of the source-water contaminant 1,1-dichloropropene: activation by GSTT1-1 and structure-activity considerations. Water 42-47 glutathione S-transferase theta 1 Homo sapiens 95-102 16161713-9 2005 The limited stratum-specific numbers showed that the exposed workers with the GSTM1-/GSTT1-genotype had nonsignificantly higher frequencies of all the effect parameters than the unexposed workers; this finding indicates that individual susceptibility related to the detoxification of acrylamide and N-methylolacrylamide may have played a role in the observed effect. Acrylamide 284-294 glutathione S-transferase theta 1 Homo sapiens 85-90 16161713-9 2005 The limited stratum-specific numbers showed that the exposed workers with the GSTM1-/GSTT1-genotype had nonsignificantly higher frequencies of all the effect parameters than the unexposed workers; this finding indicates that individual susceptibility related to the detoxification of acrylamide and N-methylolacrylamide may have played a role in the observed effect. N-methylolacrylamide 299-319 glutathione S-transferase theta 1 Homo sapiens 85-90 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Benzene 34-41 glutathione S-transferase theta 1 Homo sapiens 66-71 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Benzene 34-41 glutathione S-transferase theta 1 Homo sapiens 201-206 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Creatinine 109-119 glutathione S-transferase theta 1 Homo sapiens 66-71 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Benzene 124-131 glutathione S-transferase theta 1 Homo sapiens 66-71 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Creatinine 231-241 glutathione S-transferase theta 1 Homo sapiens 66-71 15935803-6 2005 The production rate of S-PMA from benzene in exposed workers with GSTT1 null alleles (24.72+/-32.48 microg/g creatinine/ppm benzene) was significantly lower than that in subjects with the wild type of GSTT1 (59.84+/-47.66 microg/g creatinine/ppm benzene, p<0.0001). Benzene 124-131 glutathione S-transferase theta 1 Homo sapiens 66-71 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Benzene 100-107 glutathione S-transferase theta 1 Homo sapiens 56-61 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Benzene 100-107 glutathione S-transferase theta 1 Homo sapiens 169-174 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Cotinine 122-130 glutathione S-transferase theta 1 Homo sapiens 56-61 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Cotinine 122-130 glutathione S-transferase theta 1 Homo sapiens 169-174 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Benzene 268-275 glutathione S-transferase theta 1 Homo sapiens 56-61 15935803-7 2005 Further regression analysis of S-PMA production rate on GSTT1 genotype with adjustment of sex, age, benzene exposure, and cotinine levels indicated that the genotype of GSTT1 plays a critical role in determining the inter-individual variations of S-PMA formation from benzene exposure. Benzene 268-275 glutathione S-transferase theta 1 Homo sapiens 169-174 15935803-8 2005 Therefore, the individual genotype of GSTT1 needs to be identified and considered while using S-PMA as a marker to estimate the personal exposure levels of benzene in future population studies. Benzene 156-163 glutathione S-transferase theta 1 Homo sapiens 38-43 15790493-0 2005 Genotoxicity and metabolism of the source-water contaminant 1,1-dichloropropene: activation by GSTT1-1 and structure-activity considerations. 1,1-dichloropropene 60-79 glutathione S-transferase theta 1 Homo sapiens 95-102 15748501-8 2005 The genetic polymorphisms of 3 genes (NQO1, GSTT1 and GSTM1) led to declining of detoxifying ability in benzene metabolism, so the individual with NQO1 C609T T/T genotype, GSTT1 null genotype and GSTM1 null genotype is most susceptive to benzene. Benzene 104-111 glutathione S-transferase theta 1 Homo sapiens 44-49 15668107-9 2005 In this experiment individuals carrying GSTT1 showed lower adduct level increments from ethylene oxide than individuals lacking GSTT1. Ethylene Oxide 88-102 glutathione S-transferase theta 1 Homo sapiens 40-45 15748501-8 2005 The genetic polymorphisms of 3 genes (NQO1, GSTT1 and GSTM1) led to declining of detoxifying ability in benzene metabolism, so the individual with NQO1 C609T T/T genotype, GSTT1 null genotype and GSTM1 null genotype is most susceptive to benzene. Benzene 104-111 glutathione S-transferase theta 1 Homo sapiens 172-177 15748501-8 2005 The genetic polymorphisms of 3 genes (NQO1, GSTT1 and GSTM1) led to declining of detoxifying ability in benzene metabolism, so the individual with NQO1 C609T T/T genotype, GSTT1 null genotype and GSTM1 null genotype is most susceptive to benzene. Benzene 238-245 glutathione S-transferase theta 1 Homo sapiens 44-49 15748501-8 2005 The genetic polymorphisms of 3 genes (NQO1, GSTT1 and GSTM1) led to declining of detoxifying ability in benzene metabolism, so the individual with NQO1 C609T T/T genotype, GSTT1 null genotype and GSTM1 null genotype is most susceptive to benzene. Benzene 238-245 glutathione S-transferase theta 1 Homo sapiens 172-177 16438295-5 2005 The dose of acrylamide or glycidamide has been measured in blood samples from individuals with defined genotypes for the glutathione transferases GSTT1 and GSTM1 after in vitro incubation with these compounds. Acrylamide 12-22 glutathione S-transferase theta 1 Homo sapiens 146-151 16438295-5 2005 The dose of acrylamide or glycidamide has been measured in blood samples from individuals with defined genotypes for the glutathione transferases GSTT1 and GSTM1 after in vitro incubation with these compounds. glycidamide 26-37 glutathione S-transferase theta 1 Homo sapiens 146-151 15256483-5 2004 Therefore, we hypothesized that individuals possessing the low activity genotypes of GSTM1, GSTT1 and/or GSTP1 (i.e. the GSTM1 null, GSTT1 null and GSTP1 AB/BB genotypes, respectively) may exhibit a stronger marine n-3 fatty acid-breast cancer association than their high activity counterparts. Fatty Acids, Omega-3 215-229 glutathione S-transferase theta 1 Homo sapiens 92-97 15638917-0 2005 Genetic polymorphism of glutathione S-transferase genes (GSTM1, GSTT1 and GSTP1) and susceptibility to prostate cancer in Northern India. Glutathione 24-35 glutathione S-transferase theta 1 Homo sapiens 64-69 15533900-11 2004 Subjects carrying the GSTT1 wild-type excreted higher concentrations of S-PMA than subjects carrying the null genotype, suggesting that it is a key enzyme in the glutathione conjugation that leads to S-PMA. Glutathione 162-173 glutathione S-transferase theta 1 Homo sapiens 22-27 15934438-2 2005 Glutathione S-transferase (GST) is involved in the detoxification of ROS and genetic polymorphisms of GSTM1, GSTT1 and GSTP1 are associated with altered enzyme activity. Reactive Oxygen Species 69-72 glutathione S-transferase theta 1 Homo sapiens 109-114 16521944-4 2005 Very important role in this process is played by S-glutathione transferase M1 (GSTM1) and S-glutathione transferase T1 (GSTT1) which conjugate glutathione with xenobiotics and promote their removal from human body. Glutathione 51-62 glutathione S-transferase theta 1 Homo sapiens 120-125 16521944-7 2005 The common deletions of GSTM1 and GSTT1 were determined by polymerase chain reaction and agarose gel separation. Sepharose 89-96 glutathione S-transferase theta 1 Homo sapiens 34-39 15279829-11 2004 Reaction of the dihalo and monohalo HNMs with GSH, possibly GSTT1-1, is a possible mechanism for formation of ultimate mutagenic products. Glutathione 46-49 glutathione S-transferase theta 1 Homo sapiens 60-67 15298956-0 2004 Benzo(a)pyrene diolepoxide (BPDE)-DNA adduct levels in leukocytes of smokers in relation to polymorphism of CYP1A1, GSTM1, GSTP1, GSTT1, and mEH. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 0-26 glutathione S-transferase theta 1 Homo sapiens 130-135 15298956-0 2004 Benzo(a)pyrene diolepoxide (BPDE)-DNA adduct levels in leukocytes of smokers in relation to polymorphism of CYP1A1, GSTM1, GSTP1, GSTT1, and mEH. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 28-32 glutathione S-transferase theta 1 Homo sapiens 130-135 15338373-0 2004 Genetic analysis of the glutathione s-transferase genes MGST1, GSTM3, GSTT1, and GSTM1 in patients with hereditary pancreatitis. Glutathione 24-35 glutathione S-transferase theta 1 Homo sapiens 70-75 15764300-2 2004 A preliminary case-control study was conducted to explore the association between genetic polymorphisms of GSTT1, p53 codon 72 and bladder cancer in southern Taiwan, a former high arsenic exposure area. Arsenic 180-187 glutathione S-transferase theta 1 Homo sapiens 107-112 15213713-2 2004 Functional polymorphisms in DNA-repair genes XPD, ERCC1, XRCC1, XPA, and metabolising genes glutathione S-transferase GSTP1, GSTT1, GSTM1, and thymidylate synthase (TS) were assessed retrospectively in 106 patients with refractory stage IV disease who received 5-FU/oxaliplatin combination chemotherapy, using a polymerase chain reaction-based RFLP technique. Fluorouracil 261-265 glutathione S-transferase theta 1 Homo sapiens 125-130 15213713-2 2004 Functional polymorphisms in DNA-repair genes XPD, ERCC1, XRCC1, XPA, and metabolising genes glutathione S-transferase GSTP1, GSTT1, GSTM1, and thymidylate synthase (TS) were assessed retrospectively in 106 patients with refractory stage IV disease who received 5-FU/oxaliplatin combination chemotherapy, using a polymerase chain reaction-based RFLP technique. Oxaliplatin 266-277 glutathione S-transferase theta 1 Homo sapiens 125-130 15036125-0 2004 Influence of GSTT1, mEH, CYP2E1 and RAD51 polymorphisms on diepoxybutane-induced SCE frequency in cultured human lymphocytes. diepoxybutane 59-72 glutathione S-transferase theta 1 Homo sapiens 13-18 15103050-9 2004 At 1 mM glutathione and 300 microM MeP concentrations, hGSTT1-1 and hGSTA1-1 exhibited the highest O-dealkylation activities: 545.8 and 65.0 nmol/min/mg, respectively. Glutathione 8-19 glutathione S-transferase theta 1 Homo sapiens 55-63 15256146-3 2004 RESULTS: The frequency of non-null GSTT1 gene in benzene poisoning workers with moderate benzene exposure level was higher than that in cases with lower benzene exposure (68.63% vs 38.00%, OR(adj) = 4.32, 95% CI 1.75 - 10.66, P = 0.002). Benzene 49-56 glutathione S-transferase theta 1 Homo sapiens 35-40 15256146-3 2004 RESULTS: The frequency of non-null GSTT1 gene in benzene poisoning workers with moderate benzene exposure level was higher than that in cases with lower benzene exposure (68.63% vs 38.00%, OR(adj) = 4.32, 95% CI 1.75 - 10.66, P = 0.002). Benzene 89-96 glutathione S-transferase theta 1 Homo sapiens 35-40 15256146-3 2004 RESULTS: The frequency of non-null GSTT1 gene in benzene poisoning workers with moderate benzene exposure level was higher than that in cases with lower benzene exposure (68.63% vs 38.00%, OR(adj) = 4.32, 95% CI 1.75 - 10.66, P = 0.002). Benzene 89-96 glutathione S-transferase theta 1 Homo sapiens 35-40 15256146-6 2004 CONCLUSION: There is interaction between the polymorphism of GSTT1 gene and moderate benzene exposure level; non-null GSTM1 gene and drinking x exposure level increase the risk of occupational chronic benzene poisoning; polymorphism of NQO1 gene C.609 also interacts with drinking, while polymorphism of NQO1 gene and drinking x smoking may further increase the risk of occupational chronic benzene poisoning. Benzene 85-92 glutathione S-transferase theta 1 Homo sapiens 61-66 15256146-6 2004 CONCLUSION: There is interaction between the polymorphism of GSTT1 gene and moderate benzene exposure level; non-null GSTM1 gene and drinking x exposure level increase the risk of occupational chronic benzene poisoning; polymorphism of NQO1 gene C.609 also interacts with drinking, while polymorphism of NQO1 gene and drinking x smoking may further increase the risk of occupational chronic benzene poisoning. Benzene 201-208 glutathione S-transferase theta 1 Homo sapiens 61-66 15256146-6 2004 CONCLUSION: There is interaction between the polymorphism of GSTT1 gene and moderate benzene exposure level; non-null GSTM1 gene and drinking x exposure level increase the risk of occupational chronic benzene poisoning; polymorphism of NQO1 gene C.609 also interacts with drinking, while polymorphism of NQO1 gene and drinking x smoking may further increase the risk of occupational chronic benzene poisoning. Benzene 201-208 glutathione S-transferase theta 1 Homo sapiens 61-66 15033463-1 2004 In order to find the effect of genetic polymorphisms of GSTM1 and GSTT1 on blood pressure of individuals chronically exposed to sulfur compounds, the present study was done. Sulfur 128-134 glutathione S-transferase theta 1 Homo sapiens 66-71 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. Styrene 40-47 glutathione S-transferase theta 1 Homo sapiens 117-122 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. Epoxy Compounds 55-62 glutathione S-transferase theta 1 Homo sapiens 117-122 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. 1,3-butadiene 78-91 glutathione S-transferase theta 1 Homo sapiens 117-122 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. Acetylcysteine 181-198 glutathione S-transferase theta 1 Homo sapiens 117-122 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. 1,3-butadiene 82-91 glutathione S-transferase theta 1 Homo sapiens 117-122 15093278-10 2004 For instance, in vitro SCE induction by styrene and by epoxide metabolites of 1,3-butadiene is modified by GSTM1 and GSTT1 genotypes--which also influence the excretion of specific mercapturic acids in humans exposed to butadiene and styrene. Styrene 234-241 glutathione S-transferase theta 1 Homo sapiens 117-122 15047208-9 2004 Although the frequency of overall GSTT1 null genotype was significantly lower in cervical carcinoma patients with high-risk HPV infection (OR = 0.3, 95% CI: 0.1-1.0), almost 2-fold increased risk was observed among women with GSTT1 null and Arg/Arg genotype (OR = 1.9, 95% CI: 0.7-5.4). Arginine 241-244 glutathione S-transferase theta 1 Homo sapiens 34-39 15047208-9 2004 Although the frequency of overall GSTT1 null genotype was significantly lower in cervical carcinoma patients with high-risk HPV infection (OR = 0.3, 95% CI: 0.1-1.0), almost 2-fold increased risk was observed among women with GSTT1 null and Arg/Arg genotype (OR = 1.9, 95% CI: 0.7-5.4). Arginine 245-248 glutathione S-transferase theta 1 Homo sapiens 34-39 15141365-6 2004 In the present study, we have evaluated the effect of GSTM1, GSTT1, and GSTP1 polymorphisms on the frequency of MN and SCE induced by hydroquinone in human lymphocytes. hydroquinone 134-146 glutathione S-transferase theta 1 Homo sapiens 61-66 15055296-10 2004 These findings may be explained by reduced detoxification capacity rendered by the altered gene and may be linked with exposure to, for example, heterocyclic amines in the case of NAT2 and endogenously formed ethylene oxide in the case of GSTT1. Ethylene Oxide 209-223 glutathione S-transferase theta 1 Homo sapiens 239-244 14751678-0 2004 Occupational exposure to styrene: modulation of cytogenetic damage and levels of urinary metabolites of styrene by polymorphisms in genes CYP2E1, EPHX1, GSTM1, GSTT1 and GSTP1. Styrene 25-32 glutathione S-transferase theta 1 Homo sapiens 160-165 15055296-10 2004 These findings may be explained by reduced detoxification capacity rendered by the altered gene and may be linked with exposure to, for example, heterocyclic amines in the case of NAT2 and endogenously formed ethylene oxide in the case of GSTT1. heterocyclic amines 145-164 glutathione S-transferase theta 1 Homo sapiens 239-244 12732844-10 2003 CONCLUSIONS: The double null mutation of GSTT1 and GSTM1 might influence troglitazone-associated abnormal increases of liver enzyme levels. Troglitazone 73-85 glutathione S-transferase theta 1 Homo sapiens 41-46 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. Epoxy Compounds 70-77 glutathione S-transferase theta 1 Homo sapiens 142-147 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. 1,3-butadiene 92-105 glutathione S-transferase theta 1 Homo sapiens 142-147 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. 3,4-epoxy-1-butene 256-274 glutathione S-transferase theta 1 Homo sapiens 142-147 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. Epoxy Compounds 284-291 glutathione S-transferase theta 1 Homo sapiens 142-147 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. 1,3-butadiene 306-319 glutathione S-transferase theta 1 Homo sapiens 142-147 12896903-9 2003 In HT29, butyrate significantly enhanced GSTA1/2 (3.5-fold), GSTM2 (not detectable in controls), GSTP1 (1.5-fold) and GST activity (1.4-fold), but not GSTM1 or GSTT1. Butyrates 9-17 glutathione S-transferase theta 1 Homo sapiens 160-165 14669454-0 2003 [Manifestation of glutathione S-transferase GSTM1 and GSTT1 in female patients with bleomycin-positive chromosome instability]. Bleomycin 84-93 glutathione S-transferase theta 1 Homo sapiens 54-59 14560658-4 2003 An example of multiplex PCR-CTPP is described for NQO1 C609, GSTM1, and GSTT1. ctpp 28-32 glutathione S-transferase theta 1 Homo sapiens 72-77 14582249-4 2003 Particularly the role of GSTT1 and GSTM1 polymorphism on the biological indicators of 1,3-butadiene has been evaluated. 1,3-butadiene 86-99 glutathione S-transferase theta 1 Homo sapiens 25-30 12872524-18 2003 The human polymorphic GST catalysing conjugation of halomethanes, dihalomethanes, ethylene oxide and a number of other industrial compounds could be characterised as a class theta enzyme (GSTT1) by means of molecular biology. halomethanes 52-64 glutathione S-transferase theta 1 Homo sapiens 188-193 12872524-18 2003 The human polymorphic GST catalysing conjugation of halomethanes, dihalomethanes, ethylene oxide and a number of other industrial compounds could be characterised as a class theta enzyme (GSTT1) by means of molecular biology. dihalomethanes 66-80 glutathione S-transferase theta 1 Homo sapiens 188-193 12872524-18 2003 The human polymorphic GST catalysing conjugation of halomethanes, dihalomethanes, ethylene oxide and a number of other industrial compounds could be characterised as a class theta enzyme (GSTT1) by means of molecular biology. Ethylene Oxide 82-96 glutathione S-transferase theta 1 Homo sapiens 188-193 12872524-21 2003 Human phenotyping is facilitated by the GST activity towards methyl bromide or ethylene oxide in erythrocytes which is representative of the metabolic GSTT1 competence of the entire organism. methyl bromide 61-75 glutathione S-transferase theta 1 Homo sapiens 151-156 12872524-21 2003 Human phenotyping is facilitated by the GST activity towards methyl bromide or ethylene oxide in erythrocytes which is representative of the metabolic GSTT1 competence of the entire organism. Ethylene Oxide 79-93 glutathione S-transferase theta 1 Homo sapiens 151-156 14644336-6 2003 In vitro sensitivity to the genotoxicity of 1,2:3,4-diepoxybutane, an epoxide metabolite of 1,3-butadiene has clearly been shown to depend on GSTT1 genotype, which has also been implicated to modify, along with GSTM1 genotype, the in vitro genotoxicity of 1,2-epoxy-3-butene, another epoxide metabolite of 1,3-butadiene. diepoxybutane 44-65 glutathione S-transferase theta 1 Homo sapiens 142-147 12739102-1 2003 Vinyl chloride monomer (VCM) is a known human carcinogen, which may be metabolized by cytochrome P450 2E1 (CYP2E1), aldehyde dehydrogenase 2 (ALDH2), and glutathione S-transferase T1 (GSTT1). Vinyl Chloride 0-14 glutathione S-transferase theta 1 Homo sapiens 154-182 12739102-1 2003 Vinyl chloride monomer (VCM) is a known human carcinogen, which may be metabolized by cytochrome P450 2E1 (CYP2E1), aldehyde dehydrogenase 2 (ALDH2), and glutathione S-transferase T1 (GSTT1). Vinyl Chloride 0-14 glutathione S-transferase theta 1 Homo sapiens 184-189 12916871-0 2003 Mutant type glutathione S-transferase theta 1 gene homologue to mTOR in myelodysplastic syndrome: possible clinical application of rapamycin. Sirolimus 131-140 glutathione S-transferase theta 1 Homo sapiens 12-45 12916871-7 2003 Interestingly, rapamycin could induce significant growth inhibition of the stable transformants for mutant type GSTT-1, which was indicative of apoptosis, but not that of those for wild type GSTT-1. Sirolimus 15-24 glutathione S-transferase theta 1 Homo sapiens 112-118 12916871-8 2003 These results suggest that rapamycin could be included in the therapeutic modality for the patients with MDS who have the mTOR sequences in GSTT-1 gene. Sirolimus 27-36 glutathione S-transferase theta 1 Homo sapiens 140-146 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 90-95 glutathione S-transferase theta 1 Homo sapiens 53-58 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 90-95 glutathione S-transferase theta 1 Homo sapiens 113-118 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 90-95 glutathione S-transferase theta 1 Homo sapiens 113-118 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 176-181 glutathione S-transferase theta 1 Homo sapiens 53-58 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 176-181 glutathione S-transferase theta 1 Homo sapiens 113-118 12807751-5 2003 After adjusting for these factors, we found that the GSTT1 genotypes affected the urinary 1-OHP levels, i.e. the GSTT1 present subjects had approximately 1.5 times the urinary 1-OHP level than the GSTT1 null subjects (P < 0.05). Oxaliplatin 176-181 glutathione S-transferase theta 1 Homo sapiens 113-118 12807751-6 2003 In the case of the subjects who were also GSTM1 null, this trend became stronger, i.e. the GSTT1 present subjects had approximately 2 times the urinary 1-OHP level (P < 0.01). Oxaliplatin 152-157 glutathione S-transferase theta 1 Homo sapiens 91-96 12807751-8 2003 Therefore, this study suggests that the GSTT1 genetic polymorphism has the potential to affect the biological monitoring of PAHs with urinary 1-OHP, and might act as a genetic factor in PAH-related toxicity. Oxaliplatin 142-147 glutathione S-transferase theta 1 Homo sapiens 40-45 12807751-8 2003 Therefore, this study suggests that the GSTT1 genetic polymorphism has the potential to affect the biological monitoring of PAHs with urinary 1-OHP, and might act as a genetic factor in PAH-related toxicity. Polycyclic Aromatic Hydrocarbons 124-127 glutathione S-transferase theta 1 Homo sapiens 40-45 12851839-3 2003 METHODS: In the present study we analyzed GSTM1 and GSTT1 polymorphisms in the genomic DNA isolated from peripheral blood of patients with ovarian cancer treated with chemotherapy (paclitaxel and cisplatinum) after cytoreductive surgery and assessed its correlation with the clinical outcome of these patients. Paclitaxel 181-191 glutathione S-transferase theta 1 Homo sapiens 52-57 12563680-2 2003 The glutathione S-transferase (GST) genes GSTM1, GSTT1, and GSTP1 catalyze metabolic pathways for the excretion of reactive oxygen species that may be generated by cellular oxidative stress induced by ultraviolet radiation in sunlight. Reactive Oxygen Species 115-138 glutathione S-transferase theta 1 Homo sapiens 49-54 12868187-1 2003 The aim of the study was an assessment of various risk factors for nephrotoxicity of ifosfamide (IF) in children taking into account the importance of the concentrations of toxic metabolites of the drug excreted with urine and the polymorphism of genes encoding S-glutathione transferases of mi, pi, and theta classes (GSTM1, GSTP1 and GSTT1). Ifosfamide 85-95 glutathione S-transferase theta 1 Homo sapiens 336-341 12717779-10 2003 The field survey confirms that styrene exposure is associated with increased DNA damage and indicates a modulating role for GSTM1 and GSTT1 genotypes. Styrene 31-38 glutathione S-transferase theta 1 Homo sapiens 134-139 12718704-3 2003 This paper introduces triplex PCR-CTPP to simultaneously genotype three functional polymorphisms of carcinogen-detoxifying enzymes, NQO1 C609T, GSTM1 null, and GSTT1 null, all of which are reported to have a significant association with smoking-related cancers. ctpp 34-38 glutathione S-transferase theta 1 Homo sapiens 160-165 15258326-2 2003 It catalyzes the reduction of glutathione to its thioester; thus, deficiency in GST activity due to homozygous deletion of the GSTT1 gene (null genotype) may play a role in the induction of lung cancer by smoking. Glutathione 30-41 glutathione S-transferase theta 1 Homo sapiens 127-132 20021156-10 2003 The genotype status of GSTM1 and GSTT1 was successfully determined in all patients by analyzing an aliquot of the same DNA used for the 8-OHdG evaluation. 8-ohdg 136-142 glutathione S-transferase theta 1 Homo sapiens 33-38 12460800-1 2002 Metabolic enzymes involved in benzene activation or detoxification, including NAD(P)H, quinone oxidoreductase 1 (NQO1), cytochrome P450 2E1 (CYP2E1), myeloperoxidase (MPO), glutathione-S-transferase mu-1 (GSTM1), and glutathione-S-transferase theta-1 (GSTT1), were studied for their roles in human susceptibility to benzene poisoning. Benzene 30-37 glutathione S-transferase theta 1 Homo sapiens 217-250 12460800-0 2002 Association of genetic polymorphisms in CYP2E1, MPO, NQO1, GSTM1, and GSTT1 genes with benzene poisoning. Benzene 87-94 glutathione S-transferase theta 1 Homo sapiens 70-75 12460800-1 2002 Metabolic enzymes involved in benzene activation or detoxification, including NAD(P)H, quinone oxidoreductase 1 (NQO1), cytochrome P450 2E1 (CYP2E1), myeloperoxidase (MPO), glutathione-S-transferase mu-1 (GSTM1), and glutathione-S-transferase theta-1 (GSTT1), were studied for their roles in human susceptibility to benzene poisoning. Benzene 30-37 glutathione S-transferase theta 1 Homo sapiens 252-257 12460800-7 2002 Our data also indicated that individuals with CYP2E1 c.-1293 C/C and c.-1293 G/C, and NQO1 c.609 T/T, and GSTT1 null genotypes tended to be more susceptible to benzene toxicity. Benzene 160-167 glutathione S-transferase theta 1 Homo sapiens 106-111 12460800-8 2002 Our results suggest that the combined effect of polymorphisms in NQO1, CYP2E1, and GSTT1 genes and lifestyle factors might contribute to benzene poisoning. Benzene 137-144 glutathione S-transferase theta 1 Homo sapiens 83-88 12415426-2 2002 We have developed a routine ex vivo photometric phenotyping procedure based on the determination of bromide release rates from the hGSTT1-1-catalyzed glutathione conjugation of the substrate methyl bromide in EDTA blood samples under standard conditions (1,000 ppm methyl bromide, 10 min incubation). Bromides 100-107 glutathione S-transferase theta 1 Homo sapiens 131-137 12552997-1 2002 N-acetyltransferase-2 (NAT-2) and Glutathione-S-transferase M1 and T1 (GSTM1 and GSTT1) polymorphism have been implicated in the detoxification of urothelial carcinogens, such as arylamines and polycyclic aromatic hydrocarbons. aniline 179-189 glutathione S-transferase theta 1 Homo sapiens 81-86 12552997-1 2002 N-acetyltransferase-2 (NAT-2) and Glutathione-S-transferase M1 and T1 (GSTM1 and GSTT1) polymorphism have been implicated in the detoxification of urothelial carcinogens, such as arylamines and polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 194-226 glutathione S-transferase theta 1 Homo sapiens 81-86 12415426-2 2002 We have developed a routine ex vivo photometric phenotyping procedure based on the determination of bromide release rates from the hGSTT1-1-catalyzed glutathione conjugation of the substrate methyl bromide in EDTA blood samples under standard conditions (1,000 ppm methyl bromide, 10 min incubation). Glutathione 150-161 glutathione S-transferase theta 1 Homo sapiens 131-137 12415426-2 2002 We have developed a routine ex vivo photometric phenotyping procedure based on the determination of bromide release rates from the hGSTT1-1-catalyzed glutathione conjugation of the substrate methyl bromide in EDTA blood samples under standard conditions (1,000 ppm methyl bromide, 10 min incubation). methyl bromide 191-205 glutathione S-transferase theta 1 Homo sapiens 131-137 12415426-2 2002 We have developed a routine ex vivo photometric phenotyping procedure based on the determination of bromide release rates from the hGSTT1-1-catalyzed glutathione conjugation of the substrate methyl bromide in EDTA blood samples under standard conditions (1,000 ppm methyl bromide, 10 min incubation). Edetic Acid 209-213 glutathione S-transferase theta 1 Homo sapiens 131-137 12415426-2 2002 We have developed a routine ex vivo photometric phenotyping procedure based on the determination of bromide release rates from the hGSTT1-1-catalyzed glutathione conjugation of the substrate methyl bromide in EDTA blood samples under standard conditions (1,000 ppm methyl bromide, 10 min incubation). methyl bromide 265-279 glutathione S-transferase theta 1 Homo sapiens 131-137 12172927-3 2002 Although CYP and GST enzymes are involved in the activation and detoxification of N-nitrosamines and related compound, studies on the relationship between genetic polymorphisms of CYP2E1, GSTT1, and GSTM1 and the risk of gastric carcinoma (GC) are few, and the results have been conflicting. n-nitrosamines 82-96 glutathione S-transferase theta 1 Homo sapiens 188-193 12376511-8 2002 Higher THBVal levels were found in subjects with GSTM1 null and GSTT1 null genotypes; borderline influences were also noticed for CYP2E1(G(-35)T). thbval 7-13 glutathione S-transferase theta 1 Homo sapiens 64-69 12376511-10 2002 A multiple linear regression analysis, where each factor contributed significantly, correlated THBVal levels with smoking, CYP2E1(G(-35)T), GSTT1, and GSTM1 genotypes (r = 0.698). thbval 95-101 glutathione S-transferase theta 1 Homo sapiens 140-145 14694720-0 2002 [Relation of genetic polymorphism of NQO1 and GSTT1 with risks of chronic benzene poisoning]. Benzene 74-81 glutathione S-transferase theta 1 Homo sapiens 46-51 14694720-1 2002 OBJECTIVE: To explore the relation between genetic polymorphisms of NQO1, GSTT1 and risks of chronic benzene poisoning (BP). Benzene 101-108 glutathione S-transferase theta 1 Homo sapiens 74-79 14694720-1 2002 OBJECTIVE: To explore the relation between genetic polymorphisms of NQO1, GSTT1 and risks of chronic benzene poisoning (BP). Benzo(a)pyrene 120-122 glutathione S-transferase theta 1 Homo sapiens 74-79 12055050-0 2002 GSTT1 and CYP2E1 polymorphisms and trihalomethanes in drinking water: effect on childhood leukemia. Drinking Water 54-68 glutathione S-transferase theta 1 Homo sapiens 0-5 12183419-11 2002 Analyses of combined genotypes suggested an interaction between XRCC1 (Gln/Gln or Arg/Gln) and GSTT1/GSTM1-null/null among women but not among men. Glutamine 71-74 glutathione S-transferase theta 1 Homo sapiens 95-100 12183419-11 2002 Analyses of combined genotypes suggested an interaction between XRCC1 (Gln/Gln or Arg/Gln) and GSTT1/GSTM1-null/null among women but not among men. Glutamine 75-78 glutathione S-transferase theta 1 Homo sapiens 95-100 12183419-11 2002 Analyses of combined genotypes suggested an interaction between XRCC1 (Gln/Gln or Arg/Gln) and GSTT1/GSTM1-null/null among women but not among men. Arginine 82-85 glutathione S-transferase theta 1 Homo sapiens 95-100 12183419-11 2002 Analyses of combined genotypes suggested an interaction between XRCC1 (Gln/Gln or Arg/Gln) and GSTT1/GSTM1-null/null among women but not among men. Glutamine 75-78 glutathione S-transferase theta 1 Homo sapiens 95-100 12163326-4 2002 The NQO1 and GSTT1 polymorphisms were associated with lung adenocarcinoma risk with adjusted odds ratio of 2.15 for the NQO1-Pro/Pro genotype versus the Ser/Ser genotype and adjusted odds ratio of 1.61 for the GSTT1-null genotype versus the positive genotype, respectively. Serine 153-156 glutathione S-transferase theta 1 Homo sapiens 13-18 12163326-4 2002 The NQO1 and GSTT1 polymorphisms were associated with lung adenocarcinoma risk with adjusted odds ratio of 2.15 for the NQO1-Pro/Pro genotype versus the Ser/Ser genotype and adjusted odds ratio of 1.61 for the GSTT1-null genotype versus the positive genotype, respectively. Serine 157-160 glutathione S-transferase theta 1 Homo sapiens 13-18 14992466-8 2002 An increased binomial regression risk ratio = 2.5, 95% confidence interval (CI) 1.5-4.2, of the GSTM1 null genotype for CTE was found in smokers and for the GSTT1 null genotype (binomial regression risk ratio 1.5, 95% CI 1.0-2.0). 1,1,1-trifluoro-2-chloroethane 120-123 glutathione S-transferase theta 1 Homo sapiens 157-162 12055050-2 2002 We included a subset of cases from a population-based case-control study in a case-only study to estimate the interaction odds ratios (IORs) between prenatal and postnatal exposure to THMs and polymorphisms in the GSTT1 and CYP2E1 genes. Trihalomethanes 184-188 glutathione S-transferase theta 1 Homo sapiens 214-219 12055050-4 2002 The IOR for a postnatal average of total THM above the 95th percentile with GSTT1 null genotype was 9.1 [95% confidence interval (95% CI), 1.4-57.8]. Trihalomethanes 41-44 glutathione S-transferase theta 1 Homo sapiens 76-81 12016165-9 2002 Our results suggest that the concurrent lack of the GSTM1 and GSTT1 genes increases the genotoxic effects of styrene in human cells. Styrene 109-116 glutathione S-transferase theta 1 Homo sapiens 62-67 12034316-1 2002 The aim of this study was to use DNA adducts levels, detected by 32P-postlabelling, as a biomarker to assess human exposure to polycyclic aromatic hydrocarbons (PAHs) from a coke oven plant and explore the possible association between CYP1A1 MspI, GSTP1, GSTM1 and GSTT1 genotypes, and smoking status on bulky DNA adduct formation. Polycyclic Aromatic Hydrocarbons 161-165 glutathione S-transferase theta 1 Homo sapiens 265-270 12016165-0 2002 Influence of GSTM1 and GSTT1 genotypes on sister chromatid exchange induction by styrene in cultured human lymphocytes. Styrene 81-88 glutathione S-transferase theta 1 Homo sapiens 23-28 12016165-2 2002 We previously observed that GSTT1 null genotype was associated with increased induction of sister chromatid exchanges (SCEs) by a metabolite of styrene, styrene-7,8-oxide, in human lymphocyte cultures, while GSTM1 genotype had no effect. Styrene 144-151 glutathione S-transferase theta 1 Homo sapiens 28-33 12016165-2 2002 We previously observed that GSTT1 null genotype was associated with increased induction of sister chromatid exchanges (SCEs) by a metabolite of styrene, styrene-7,8-oxide, in human lymphocyte cultures, while GSTM1 genotype had no effect. styrene oxide 153-170 glutathione S-transferase theta 1 Homo sapiens 28-33 12016165-6 2002 In two separate experiments, the mean number of SCEs/cell induced by 1.5 mM styrene was 1.55 times (P = 0.011) or 1.34 times (P = 0.015) higher in subjects lacking both GSTM1 and GSTT1 than in subjects having both genes. Styrene 76-83 glutathione S-transferase theta 1 Homo sapiens 179-184 12010862-11 2002 Additional analysis revealed that individuals possessing more susceptible XRCC1 Gln-Gln, CYP2E1 c2c2, ALDH2 1-2/2-2, and non-null GSTT1 genotypes were more likely to reveal p53 overexpression. Glutamine 80-83 glutathione S-transferase theta 1 Homo sapiens 130-135 11927838-2 2002 Here, we extended the study to examine the possible role of N-acetyltransferase (NAT) genotypes in the development of diisocyanate-induced ill effects, both separately and in combination with the previously examined GSTM1, GSTM3, GSTP1 and GSTT1 genotypes. 4,4'-diphenylmethane diisocyanate 118-130 glutathione S-transferase theta 1 Homo sapiens 240-245 11841793-0 2002 The polymorphic human glutathione transferase T1-1, the most efficient glutathione transferase in the denitrosation and inactivation of the anticancer drug 1,3-bis(2-chloroethyl)-1-nitrosourea. Carmustine 156-192 glutathione S-transferase theta 1 Homo sapiens 22-50 11766168-5 2001 TMA/blood benzene ratio was partially modulated by glutathione S-transferase (GST) genotypes, with significantly higher values in null individuals (GSTM1 and GSTT1 combined). muconic acid 0-3 glutathione S-transferase theta 1 Homo sapiens 158-163 11760815-0 2001 Dependence of papanicolaou gradings of exfoliated urothelial cells upon GSTM1 and GSTT1 polymorphism in benzidine-exposed workers of the Shanghai dye industry. benzidine 104-113 glutathione S-transferase theta 1 Homo sapiens 82-87 11641039-0 2001 Effects of genetic polymorphisms of CYP1A1, CYP2E1, GSTM1, and GSTT1 on the urinary levels of 1-hydroxypyrene and 2-naphthol in aircraft maintenance workers. 1-hydroxypyrene 94-109 glutathione S-transferase theta 1 Homo sapiens 63-68 11712598-5 2001 A significantly higher percentage (50%) of Cr workers had both the null GSTM1 and GSTT1 genotype as compared to 10% of the controls (P < 0.01). Chromium 43-45 glutathione S-transferase theta 1 Homo sapiens 82-87 11757669-0 2001 Polymorphisms of glutathione S-transferase genes (GSTM1, GSTP1 and GSTT1) and bladder cancer susceptibility in the Turkish population. Glutathione 17-28 glutathione S-transferase theta 1 Homo sapiens 67-72 11535247-2 2001 Previously, an extended haemoglobin adduct monitoring (N-(cyanoethyl)valine and N-(hydroxyethyl)-valine) was performed regarding the glutathione transferases hGSTM1 and hGSTT1 polymorphisms but no influence of hGSTM1 or hGSTT1 polymorphisms on specific adduct levels was found. n-(cyanoethyl)valine 55-75 glutathione S-transferase theta 1 Homo sapiens 169-175 11535247-7 2001 The study also confirmed the impact of GSTT1 polymorphism on background N-(hydroxyethyl)-valine adduct levels in haemoglobin which are caused by endogenous ethylene oxide. n-(hydroxyethyl)-valine 72-95 glutathione S-transferase theta 1 Homo sapiens 39-44 11535247-7 2001 The study also confirmed the impact of GSTT1 polymorphism on background N-(hydroxyethyl)-valine adduct levels in haemoglobin which are caused by endogenous ethylene oxide. Ethylene Oxide 156-170 glutathione S-transferase theta 1 Homo sapiens 39-44 11535248-4 2001 Vitamin C was higher in GSTT1+ compared with GSTT1 null--as was glucose-6-phosphate dehydrogenase activity. Ascorbic Acid 0-9 glutathione S-transferase theta 1 Homo sapiens 24-29 11535248-4 2001 Vitamin C was higher in GSTT1+ compared with GSTT1 null--as was glucose-6-phosphate dehydrogenase activity. Ascorbic Acid 0-9 glutathione S-transferase theta 1 Homo sapiens 45-50 11535248-9 2001 In contrast, the link between smoking and oxidised pyrimidines in DNA was seen only in the GSTT1 null group. Pyrimidines 51-62 glutathione S-transferase theta 1 Homo sapiens 91-96 11600727-10 2001 There was less association between the concentrations of 1-OHP and the GSTM1, GSTP1, or GSTT1 polymorphism. Oxaliplatin 57-62 glutathione S-transferase theta 1 Homo sapiens 88-93 11725342-11 2001 The best-known example is probably the diepoxybutane sensitivity of GSTT1 null donors. diepoxybutane 39-52 glutathione S-transferase theta 1 Homo sapiens 68-73 11641039-0 2001 Effects of genetic polymorphisms of CYP1A1, CYP2E1, GSTM1, and GSTT1 on the urinary levels of 1-hydroxypyrene and 2-naphthol in aircraft maintenance workers. 2-naphthol 114-124 glutathione S-transferase theta 1 Homo sapiens 63-68 11511185-3 2001 Activation of 1,2-dihaloethanes (BrCH(2)CH(2)Br, BrCH(2)CH(2)Cl, and ClCH(2)CH(2)Cl) was investigated using two mammalian theta class GSH transferases (rat GST 5-5 and human GST T1) and a bacterial dichloromethane dehalogenase (DM11). 1,2-dihaloethanes 14-31 glutathione S-transferase theta 1 Homo sapiens 174-180 11448648-0 2001 Host determinants of DNA alkylation and DNA repair activity in human colorectal tissue: O(6)-methylguanine levels are associated with GSTT1 genotype and O(6)-alkylguanine-DNA alkyltransferase activity with CYP2D6 genotype. O-(6)-methylguanine 89-106 glutathione S-transferase theta 1 Homo sapiens 134-139 11511186-15 2001 The halide order appears most important in the dihalomethane conjugation reactions catalyzed by GST 5-5 and less so in GST T1 and DM11, probably due to changes in the rate-limiting steps. halide 4-10 glutathione S-transferase theta 1 Homo sapiens 119-125 11446825-0 2001 A Bayesian analysis of the influence of GSTT1 polymorphism on the cancer risk estimate for dichloromethane. Methylene Chloride 91-106 glutathione S-transferase theta 1 Homo sapiens 40-45 11511186-15 2001 The halide order appears most important in the dihalomethane conjugation reactions catalyzed by GST 5-5 and less so in GST T1 and DM11, probably due to changes in the rate-limiting steps. dihalomethane 47-60 glutathione S-transferase theta 1 Homo sapiens 119-125 11418090-0 2001 Biological monitoring the exposure to polycyclic aromatic hydrocarbons of coke oven workers in relation to smoking and genetic polymorphisms for GSTM1 and GSTT1. Polycyclic Aromatic Hydrocarbons 38-70 glutathione S-transferase theta 1 Homo sapiens 155-160 11418090-3 2001 OBJECTIVE: This study aimed to assess whether the current exposure to PAH of coke oven workers in a Dutch plant induced biological effects, and to determine if these effects are influenced by tobacco smoking and by genetic polymorphisms for the glutathione S-transferase genes GSTM1 and GSTT1. Polycyclic Aromatic Hydrocarbons 70-73 glutathione S-transferase theta 1 Homo sapiens 287-292 11418090-3 2001 OBJECTIVE: This study aimed to assess whether the current exposure to PAH of coke oven workers in a Dutch plant induced biological effects, and to determine if these effects are influenced by tobacco smoking and by genetic polymorphisms for the glutathione S-transferase genes GSTM1 and GSTT1. Glutathione 245-256 glutathione S-transferase theta 1 Homo sapiens 287-292 11470760-6 2001 In addition, the effect of aflatoxin exposure on HCC risk was more pronounced among chronic HBsAg carriers with the GSTT1 null genotype (OR 3.7, 95% CI 1.5-9.3) than those who were non-null (OR 0.9, 95% CI 0.3-2.4). Aflatoxins 27-36 glutathione S-transferase theta 1 Homo sapiens 116-121 11470760-7 2001 The interaction between serum AFB(1)-albumin adduct level and GSTT1 genotype was statistically significant (P = 0.03). Aflatoxin B1 30-36 glutathione S-transferase theta 1 Homo sapiens 62-67 11446825-1 2001 The carcinogenicity of dichloromethane (DCM) is related to metabolic activation mediated by glutathione transferase theta 1 (GSTT1), whereas oxidation serves as a detoxification pathway. Methylene Chloride 23-38 glutathione S-transferase theta 1 Homo sapiens 92-123 11446825-1 2001 The carcinogenicity of dichloromethane (DCM) is related to metabolic activation mediated by glutathione transferase theta 1 (GSTT1), whereas oxidation serves as a detoxification pathway. Methylene Chloride 23-38 glutathione S-transferase theta 1 Homo sapiens 125-130 11446825-1 2001 The carcinogenicity of dichloromethane (DCM) is related to metabolic activation mediated by glutathione transferase theta 1 (GSTT1), whereas oxidation serves as a detoxification pathway. Methylene Chloride 40-43 glutathione S-transferase theta 1 Homo sapiens 92-123 11446825-1 2001 The carcinogenicity of dichloromethane (DCM) is related to metabolic activation mediated by glutathione transferase theta 1 (GSTT1), whereas oxidation serves as a detoxification pathway. Methylene Chloride 40-43 glutathione S-transferase theta 1 Homo sapiens 125-130 11397403-5 2001 While it is clear that lymphocytes from GSTT1 null individuals are more sensitive for the induction of sister chromatid exchanges (SCE) following in vitro exposure to 1,2,3,4-diepoxybutane, there was no such increase in SCE or other biomarkers of genotoxicity in workers exposed to 1-3 p.p.m. diepoxybutane 167-188 glutathione S-transferase theta 1 Homo sapiens 40-45 11556154-0 2001 Inhibition of the human erythrocytic glutathione-S-transferase T1 (GST T1) by thimerosal. Thimerosal 78-88 glutathione S-transferase theta 1 Homo sapiens 37-65 11556154-0 2001 Inhibition of the human erythrocytic glutathione-S-transferase T1 (GST T1) by thimerosal. Thimerosal 78-88 glutathione S-transferase theta 1 Homo sapiens 67-73 11556154-1 2001 We have investigated the interaction of thimerosal, a widely used antiseptic and preservative, with the human erythrocytic GST T1 (glutathione-S-transferase T1). Thimerosal 40-50 glutathione S-transferase theta 1 Homo sapiens 123-129 11556154-1 2001 We have investigated the interaction of thimerosal, a widely used antiseptic and preservative, with the human erythrocytic GST T1 (glutathione-S-transferase T1). Thimerosal 40-50 glutathione S-transferase theta 1 Homo sapiens 131-159 11556154-5 2001 For the normal conjugator we have determined a 2.77 mM thimerosal concentration to inhibit 50% of the GST T1 activity. Thimerosal 55-65 glutathione S-transferase theta 1 Homo sapiens 102-108 11556154-8 2001 Thus, sufficiently high doses of thimerosal may be able to change the phenotypic status of an individual--at least in vitro--by inhibition of the GST T1 enzyme. Thimerosal 33-43 glutathione S-transferase theta 1 Homo sapiens 146-152 11434510-5 2001 Approximately 50% of the Caucasian population are homozygous for deletions in GSTM1 and approximately 20% are homozygous for deletions in GSTT1, resulting in conjugation deficiency of mutagenic electrophiles to glutathione. Glutathione 211-222 glutathione S-transferase theta 1 Homo sapiens 138-143 11323399-0 2001 Effects of occupation, lifestyle and genetic polymorphisms of CYP1A1, CYP2E1, GSTM1 and GSTT1 on urinary 1-hydroxypyrene and 2-naphthol concentrations. 2-naphthol 125-135 glutathione S-transferase theta 1 Homo sapiens 88-93 11320155-6 2001 We have studied TSO-induced sister chromatid exchanges (SCEs) in 72 h whole-blood lymphocyte cultures from 24 healthy human donors, representing different combinations of GSTM1 and GSTT1 positive and null genotypes. stilbene oxide 16-19 glutathione S-transferase theta 1 Homo sapiens 181-186 11352866-2 2001 We examined the effects of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) genotypes on the levels of N-(2-hydroxyethyl)valine (HEV) adducts in the erythrocytes and sister chromatid exchange (SCE) in lymphocytes from a group of 58 operators of sterilizers that used EtO and nonexposed workers from nine hospitals in the United States and one hospital in Mexico City. Nitrogen 106-107 glutathione S-transferase theta 1 Homo sapiens 27-55 11352866-2 2001 We examined the effects of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) genotypes on the levels of N-(2-hydroxyethyl)valine (HEV) adducts in the erythrocytes and sister chromatid exchange (SCE) in lymphocytes from a group of 58 operators of sterilizers that used EtO and nonexposed workers from nine hospitals in the United States and one hospital in Mexico City. Nitrogen 106-107 glutathione S-transferase theta 1 Homo sapiens 57-62 11352866-2 2001 We examined the effects of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) genotypes on the levels of N-(2-hydroxyethyl)valine (HEV) adducts in the erythrocytes and sister chromatid exchange (SCE) in lymphocytes from a group of 58 operators of sterilizers that used EtO and nonexposed workers from nine hospitals in the United States and one hospital in Mexico City. 2-hydroxyethylvaline 108-130 glutathione S-transferase theta 1 Homo sapiens 57-62 11352866-2 2001 We examined the effects of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) genotypes on the levels of N-(2-hydroxyethyl)valine (HEV) adducts in the erythrocytes and sister chromatid exchange (SCE) in lymphocytes from a group of 58 operators of sterilizers that used EtO and nonexposed workers from nine hospitals in the United States and one hospital in Mexico City. 2-hydroxyethylvaline 132-135 glutathione S-transferase theta 1 Homo sapiens 27-55 11352866-2 2001 We examined the effects of glutathione S-transferase T1 (GSTT1) and M1 (GSTM1) genotypes on the levels of N-(2-hydroxyethyl)valine (HEV) adducts in the erythrocytes and sister chromatid exchange (SCE) in lymphocytes from a group of 58 operators of sterilizers that used EtO and nonexposed workers from nine hospitals in the United States and one hospital in Mexico City. 2-hydroxyethylvaline 132-135 glutathione S-transferase theta 1 Homo sapiens 57-62 11352866-7 2001 The inverse SCE-GSTT1 relationship remained unchanged when SCE was further examined in relation to HEV adducts as an indicator of the internal EtO dose. Ethylene Oxide 143-146 glutathione S-transferase theta 1 Homo sapiens 16-21 11352866-9 2001 These data indicate that the GSTT1-null genotype is associated with increased formation of EtO-hemoglobin adducts in relation to occupational EtO exposure, suggesting that individuals with homozygous deletion of the GSTT1 gene may be more susceptible to the genotoxic effects of ETO: The unexpected finding of decreased SCEs, which is less clear, may be attributed to the nonchemical specificity of this end point and the lack of expression of the GSTT1 enzyme in lymphocytes. Ethylene Oxide 279-282 glutathione S-transferase theta 1 Homo sapiens 29-34 11352866-9 2001 These data indicate that the GSTT1-null genotype is associated with increased formation of EtO-hemoglobin adducts in relation to occupational EtO exposure, suggesting that individuals with homozygous deletion of the GSTT1 gene may be more susceptible to the genotoxic effects of ETO: The unexpected finding of decreased SCEs, which is less clear, may be attributed to the nonchemical specificity of this end point and the lack of expression of the GSTT1 enzyme in lymphocytes. Ethylene Oxide 279-282 glutathione S-transferase theta 1 Homo sapiens 216-221 11352866-9 2001 These data indicate that the GSTT1-null genotype is associated with increased formation of EtO-hemoglobin adducts in relation to occupational EtO exposure, suggesting that individuals with homozygous deletion of the GSTT1 gene may be more susceptible to the genotoxic effects of ETO: The unexpected finding of decreased SCEs, which is less clear, may be attributed to the nonchemical specificity of this end point and the lack of expression of the GSTT1 enzyme in lymphocytes. Ethylene Oxide 279-282 glutathione S-transferase theta 1 Homo sapiens 216-221 11320155-0 2001 Trans-stilbene oxide-induced sister chromatid exchange in cultured human lymphocytes: influence of GSTM1 and GSTT1 genotypes. stilbene oxide 0-20 glutathione S-transferase theta 1 Homo sapiens 109-114 11323399-0 2001 Effects of occupation, lifestyle and genetic polymorphisms of CYP1A1, CYP2E1, GSTM1 and GSTT1 on urinary 1-hydroxypyrene and 2-naphthol concentrations. 1-hydroxypyrene 105-120 glutathione S-transferase theta 1 Homo sapiens 88-93 11218045-8 2001 There was little influence of known polymorphisms of GSTM1, GSTM3 and GSTP1 upon the activities towards the test substrates, whereas the influence of GSTT1 polymorphism on the activity towads methyl chloride was straightforward. Methyl Chloride 192-207 glutathione S-transferase theta 1 Homo sapiens 150-155 11305778-0 2001 High-performance liquid chromatography/fluorescence detection of S-methylglutathione formed by glutathione-S-transferase T1 in vitro. S-methyl glutathione 65-84 glutathione S-transferase theta 1 Homo sapiens 95-123 11305778-1 2001 Glutathione-S-transferase T1 (GSTT1-1) is a major isoenzyme for the biotransformation of halomethanes. halomethanes 89-101 glutathione S-transferase theta 1 Homo sapiens 0-28 11305778-1 2001 Glutathione-S-transferase T1 (GSTT1-1) is a major isoenzyme for the biotransformation of halomethanes. halomethanes 89-101 glutathione S-transferase theta 1 Homo sapiens 30-37 11305778-3 2001 Metabolism of the halomethanes via GSTT1-1 yields S-methylglutathione (MeSG). halomethanes 18-30 glutathione S-transferase theta 1 Homo sapiens 35-42 11305778-3 2001 Metabolism of the halomethanes via GSTT1-1 yields S-methylglutathione (MeSG). S-methyl glutathione 50-69 glutathione S-transferase theta 1 Homo sapiens 35-42 11305778-3 2001 Metabolism of the halomethanes via GSTT1-1 yields S-methylglutathione (MeSG). 6-mercapto-7-methylguanosine 71-75 glutathione S-transferase theta 1 Homo sapiens 35-42 11018744-15 2000 Thus, the GSTT1-1 genotype is suspected to confer decreased or increased risk of cancer in relation to the source of exposure; in vitro studies, mostly conducted on metabolites of butadiene, confirm the protective action of GSTT1-1, whereas, thus far, experimental studies prove that the increasing risk is limited. 1,3-butadiene 180-189 glutathione S-transferase theta 1 Homo sapiens 224-231 11186134-5 2000 In univariate analysis, homozygous deletion of GSTT1 (null genotype) conferred a 6.7-fold reduction in risk of prednisone poor-response compared to individuals who were either heterozygous or homozygous for GSTT1 [odds ratio (OR) = 0.15, P = 0.071; multivariate odds ratio = 0.18, P = 0.117]. Prednisone 111-121 glutathione S-transferase theta 1 Homo sapiens 47-52 23886278-1 2001 The objective of this study was to test the infiuence of genetic polymorphisms for metabolic enzymes (CYP2E1, mEH, GSTM1 and GSTT1) implicated in the biotransformation of styrene in humans on the interpretation of urinary biomarkers of exposure. Styrene 171-178 glutathione S-transferase theta 1 Homo sapiens 125-130 11159743-1 2001 The phase II glutathione S-transferases (GSTs) GSTT1, GSTM1 and GSTP1 catalyse glutathione-mediated reduction of exogenous and endogenous electrophiles. Glutathione 13-24 glutathione S-transferase theta 1 Homo sapiens 47-52 11052546-1 2000 This study investigated whether the association between low level benzene exposure and shortened gestation is modified by two susceptibility genes, CYP1A1 and GSTT1. Benzene 66-73 glutathione S-transferase theta 1 Homo sapiens 159-164 10794492-11 2000 This contrasts with a recent report of reduced risk of RCC associated with GSTT1 null in a cohort of trichloroethene-exposed workers and suggests that specific chemical exposures alter the effect of GSTT1 on cancer risk. Trichloroethylene 101-116 glutathione S-transferase theta 1 Homo sapiens 75-80 11085692-2 2000 Glutathione S-transferases M1 and T1 (GSTM1 and GSTT1) conjugate isothiocyanates leading to more rapid elimination. Isothiocyanates 65-80 glutathione S-transferase theta 1 Homo sapiens 48-53 11085692-8 2000 This protective effect of isothiocyanates was seen primarily among individuals with homozygous deletion of GSTM1 (0.36 [0.20-0.63]) and particularly with deletion of both GSTM1 and GSTT1 (0.28 [0.13-0.57]). Isothiocyanates 26-41 glutathione S-transferase theta 1 Homo sapiens 181-186 10862521-7 2000 An especially remarkable risk of breast cancer was observed for alcohol-consuming premenopausal women lacking both the GSTM1 and GSTT1 genes (OR = 5.3, 95% CI = 1.0-27.8) compared to those with both of the genes. Alcohols 64-71 glutathione S-transferase theta 1 Homo sapiens 129-134 11037805-1 2000 The aim of the present study was to investigate how the genetic polymorphism in glutathione transferase T1 (GSTT1) affects the metabolism and disposition of methyl chloride in humans in vivo. Methyl Chloride 157-172 glutathione S-transferase theta 1 Homo sapiens 108-113 11037805-2 2000 The 24 volunteers (13 males and 11 females) who participated in the study were recruited from a group of 208 individuals previously phenotyped for GSTT1 by measuring the glutathione transferase activity with methyl chloride in lysed erythrocytes ex vivo. Methyl Chloride 208-223 glutathione S-transferase theta 1 Homo sapiens 147-152 11037805-6 2000 The average net respiratory uptake of methyl chloride was 243, 158, and 44 micromol in individuals with high, intermediate and no GSTT1 activity, respectively. Methyl Chloride 38-53 glutathione S-transferase theta 1 Homo sapiens 130-135 11037805-10 2000 In conclusion, GSTT1 appears to be the sole determinant of methyl chloride metabolism in humans. Methyl Chloride 59-74 glutathione S-transferase theta 1 Homo sapiens 15-20 11037805-11 2000 Thus, individuals with nonfunctional GSTT1 entirely lack the capacity to metabolize methyl chloride. Methyl Chloride 84-99 glutathione S-transferase theta 1 Homo sapiens 37-42 11007341-11 2000 Glutathione S-transferase T1 deficiency in the thimerosal/mercury group (19.8%) was barely elevated versus healthy controls (16.0%) and the "para-compound" group (14.0%). Thimerosal 47-57 glutathione S-transferase theta 1 Homo sapiens 0-28 11007341-11 2000 Glutathione S-transferase T1 deficiency in the thimerosal/mercury group (19.8%) was barely elevated versus healthy controls (16.0%) and the "para-compound" group (14.0%). Mercury 58-65 glutathione S-transferase theta 1 Homo sapiens 0-28 10813720-6 2000 The individuals carrying both the arginine/proline genotype of p53 and the null genotype of GSTT1 showed a 3.5-fold increase of cervical carcinoma risk (95% CI, 1.8-7.1) compared with those individuals carrying both the arginine/arginine genotype of p53 and the GSTT1 positive genotype. Arginine 34-42 glutathione S-transferase theta 1 Homo sapiens 262-267 10813720-6 2000 The individuals carrying both the arginine/proline genotype of p53 and the null genotype of GSTT1 showed a 3.5-fold increase of cervical carcinoma risk (95% CI, 1.8-7.1) compared with those individuals carrying both the arginine/arginine genotype of p53 and the GSTT1 positive genotype. Arginine 220-228 glutathione S-transferase theta 1 Homo sapiens 92-97 10813720-6 2000 The individuals carrying both the arginine/proline genotype of p53 and the null genotype of GSTT1 showed a 3.5-fold increase of cervical carcinoma risk (95% CI, 1.8-7.1) compared with those individuals carrying both the arginine/arginine genotype of p53 and the GSTT1 positive genotype. Arginine 220-228 glutathione S-transferase theta 1 Homo sapiens 92-97 10813720-9 2000 CONCLUSIONS: The results of the current study suggested that the arginine/proline genotype of p53, independently or in conjunction with the GSTT1 null genotype, could affect the genetic susceptibility for cervical carcinoma, and HPV positive women carrying both null genotypes of GSTT1 and GSTM1 have an increased risk of cervical carcinoma developing before age 40 years. Arginine 65-73 glutathione S-transferase theta 1 Homo sapiens 280-285 10813720-9 2000 CONCLUSIONS: The results of the current study suggested that the arginine/proline genotype of p53, independently or in conjunction with the GSTT1 null genotype, could affect the genetic susceptibility for cervical carcinoma, and HPV positive women carrying both null genotypes of GSTT1 and GSTM1 have an increased risk of cervical carcinoma developing before age 40 years. Proline 74-81 glutathione S-transferase theta 1 Homo sapiens 280-285 10815689-1 2000 Individuals with a homozygous deletion of the glutathione S-transferase theta 1 (GSTT1) gene lack GSTT1 enzymatic detoxification of environmental carcinogens by conjugation with glutathione. Glutathione 46-57 glutathione S-transferase theta 1 Homo sapiens 81-86 10815689-1 2000 Individuals with a homozygous deletion of the glutathione S-transferase theta 1 (GSTT1) gene lack GSTT1 enzymatic detoxification of environmental carcinogens by conjugation with glutathione. Glutathione 46-57 glutathione S-transferase theta 1 Homo sapiens 98-103 10794492-11 2000 This contrasts with a recent report of reduced risk of RCC associated with GSTT1 null in a cohort of trichloroethene-exposed workers and suggests that specific chemical exposures alter the effect of GSTT1 on cancer risk. Trichloroethylene 101-116 glutathione S-transferase theta 1 Homo sapiens 199-204 10666194-0 2000 Polymorphisms within glutathione S-transferase genes (GSTM1, GSTT1, GSTP1) and risk of relapse in childhood B-cell precursor acute lymphoblastic leukemia: a case-control study. Glutathione 21-32 glutathione S-transferase theta 1 Homo sapiens 61-66 10801254-1 2000 BACKGROUND: Glutathione conjugation of tacrine reactive metabolites depends in part on the activity of glutathione-S-transferases (GST), of which two isozymes (GST M1 and GST T1) are polymorphically expressed. Glutathione 12-23 glutathione S-transferase theta 1 Homo sapiens 171-177 10801254-1 2000 BACKGROUND: Glutathione conjugation of tacrine reactive metabolites depends in part on the activity of glutathione-S-transferases (GST), of which two isozymes (GST M1 and GST T1) are polymorphically expressed. Tacrine 39-46 glutathione S-transferase theta 1 Homo sapiens 171-177 10607731-1 2000 The modulation of benzo[a]pyrene diolepoxide (BPDE)-DNA adduct levels by polymorphisms in the CYP1A1, GSTM1 and GSTT1 genes was assessed in leukocytes of Caucasian males. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 18-44 glutathione S-transferase theta 1 Homo sapiens 112-117 10667466-10 2000 After controlling for age, gender, education, pack-years of smoking, alcohol drinking, body mass index, H. pylori infection, and fruit and salt intake, the adjusted odds ratio (OR) for GSTT1 and gastric cancer was 2.50 (95% confidence interval (CI), 1.01-6.22). Alcohols 69-76 glutathione S-transferase theta 1 Homo sapiens 185-190 10667466-10 2000 After controlling for age, gender, education, pack-years of smoking, alcohol drinking, body mass index, H. pylori infection, and fruit and salt intake, the adjusted odds ratio (OR) for GSTT1 and gastric cancer was 2.50 (95% confidence interval (CI), 1.01-6.22). Salts 139-143 glutathione S-transferase theta 1 Homo sapiens 185-190 10708971-0 2000 The influence of GSTM1 and GSTT1 genotypes on the induction of sister chromatid exchanges and chromosome aberrations by 1,2:3,4-diepoxybutane. diepoxybutane 120-141 glutathione S-transferase theta 1 Homo sapiens 27-32 10607731-1 2000 The modulation of benzo[a]pyrene diolepoxide (BPDE)-DNA adduct levels by polymorphisms in the CYP1A1, GSTM1 and GSTT1 genes was assessed in leukocytes of Caucasian males. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 46-50 glutathione S-transferase theta 1 Homo sapiens 112-117 11525595-7 2000 The inverse associations between plasma selenium level and AFB1-albumin adducts were statistically significant among those with null genotypes of GSTM1 and GSTT1, but not among the nonnull genotypes. Selenium 40-48 glutathione S-transferase theta 1 Homo sapiens 156-161 10607731-0 2000 Modulation of benzo[a]pyrene diolepoxide-DNA adduct levels in human white blood cells by CYP1A1, GSTM1 and GSTT1 polymorphism. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 14-40 glutathione S-transferase theta 1 Homo sapiens 107-112 10526210-3 1999 The metabolism of brominated THMs is thought to involve a GSH conjugation reaction leading either to formaldehyde or DNA-reactive intermediates via glutathione S-transferase-theta (GSTT1-1), which is polymorphic in humans. Trihalomethanes 29-33 glutathione S-transferase theta 1 Homo sapiens 181-188 10650922-0 1999 Differential substrate behaviours of ethylene oxide and propylene oxide towards human glutathione transferase theta hGSTT1-1. Ethylene Oxide 37-51 glutathione S-transferase theta 1 Homo sapiens 116-124 10650922-0 1999 Differential substrate behaviours of ethylene oxide and propylene oxide towards human glutathione transferase theta hGSTT1-1. propylene oxide 56-71 glutathione S-transferase theta 1 Homo sapiens 116-124 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. Ethylene Oxide 22-36 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. Ethylene Oxide 22-36 glutathione S-transferase theta 1 Homo sapiens 134-139 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. Ethylene Oxide 38-40 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. propylene oxide 43-58 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. propylene oxide 60-62 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. 1,2-epoxybutane 68-84 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. 1,2-epoxybutane 68-84 glutathione S-transferase theta 1 Homo sapiens 134-139 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. 1,2-epoxybutane 86-91 glutathione S-transferase theta 1 Homo sapiens 133-141 10650922-1 1999 The transformation of ethylene oxide (EO), propylene oxide (PO) and 1-butylene oxide (1-BuO) by human glutathione transferase theta (hGSTT1-1) was studied comparatively using "conjugator" (GSTT1 + individuals) erythrocyte lysates. 1,2-epoxybutane 86-91 glutathione S-transferase theta 1 Homo sapiens 134-139 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. Epoxy Compounds 44-52 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. alkyl halides 118-131 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. methyl bromide 133-147 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. bromoethane 134-147 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. 1-bromopropane 175-191 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. 1-bromopropane 193-197 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. methyl bromide 227-231 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-4 1999 This sequence of reactivities of homologous epoxides towards GSTT1-1 contrasts to the sequence observed in homologous alkyl halides (methyl bromide, MBr; ethyl bromide, EtBr; n-propyl bromide, PrBr) where the relative sequence MeBr >> EtBr > PrBr is observed. 1-bromopropane 251-255 glutathione S-transferase theta 1 Homo sapiens 61-68 10650922-5 1999 The higher reactivity towards GSTT1-1 of propylene oxide compared to ethylene oxide is consistent with a higher chemical reactivity. propylene oxide 41-56 glutathione S-transferase theta 1 Homo sapiens 30-37 10526210-3 1999 The metabolism of brominated THMs is thought to involve a GSH conjugation reaction leading either to formaldehyde or DNA-reactive intermediates via glutathione S-transferase-theta (GSTT1-1), which is polymorphic in humans. Glutathione 58-61 glutathione S-transferase theta 1 Homo sapiens 181-188 10526210-3 1999 The metabolism of brominated THMs is thought to involve a GSH conjugation reaction leading either to formaldehyde or DNA-reactive intermediates via glutathione S-transferase-theta (GSTT1-1), which is polymorphic in humans. Formaldehyde 101-113 glutathione S-transferase theta 1 Homo sapiens 181-188 10511265-9 1999 On the other hand, 1-OHPG excretion was higher in GSTT1-positive smokers than in GSTT1-deficient smokers. 1-ohpg 19-25 glutathione S-transferase theta 1 Homo sapiens 50-55 10511265-9 1999 On the other hand, 1-OHPG excretion was higher in GSTT1-positive smokers than in GSTT1-deficient smokers. 1-ohpg 19-25 glutathione S-transferase theta 1 Homo sapiens 81-86 10511265-10 1999 It is important to note the variability of individual PAH metabolite excretion due to different GSTM1 and GSTT1 genotypes. Polycyclic Aromatic Hydrocarbons 54-57 glutathione S-transferase theta 1 Homo sapiens 106-111 10438655-1 1999 The carcinogenic potential of dichloromethane (DCM) has been linked to its metabolism to formaldehyde by glutathione-S-transferase theta 1 (GSTT1). Methylene Chloride 30-45 glutathione S-transferase theta 1 Homo sapiens 105-138 10473651-12 1999 Such results suggest a model in which exposure to brominated THMs may pose an excess genotoxic risk in GSTT1-1(+) individuals to those organs and tissues that both express this gene and come into direct contact with the brominated THM, such as the colon. Trihalomethanes 61-65 glutathione S-transferase theta 1 Homo sapiens 103-110 10473651-12 1999 Such results suggest a model in which exposure to brominated THMs may pose an excess genotoxic risk in GSTT1-1(+) individuals to those organs and tissues that both express this gene and come into direct contact with the brominated THM, such as the colon. Trihalomethanes 61-64 glutathione S-transferase theta 1 Homo sapiens 103-110 10438655-1 1999 The carcinogenic potential of dichloromethane (DCM) has been linked to its metabolism to formaldehyde by glutathione-S-transferase theta 1 (GSTT1). Methylene Chloride 30-45 glutathione S-transferase theta 1 Homo sapiens 140-145 10438655-1 1999 The carcinogenic potential of dichloromethane (DCM) has been linked to its metabolism to formaldehyde by glutathione-S-transferase theta 1 (GSTT1). Formaldehyde 89-101 glutathione S-transferase theta 1 Homo sapiens 105-138 10438655-1 1999 The carcinogenic potential of dichloromethane (DCM) has been linked to its metabolism to formaldehyde by glutathione-S-transferase theta 1 (GSTT1). Formaldehyde 89-101 glutathione S-transferase theta 1 Homo sapiens 140-145 10571654-9 1999 Based on this short review we conclude that (i) BPDE-DNA adduct levels resulting from "at risk" genotype combinations may serve as markers to identify most susceptible individuals; (ii) in Indian betel quid/tobacco chewers, the null genotypes of GSTM1 and GSTT1 greatly increased the risk for developing oral leukoplakia. 7,8-Dihydro-7,8-dihydroxybenzo(a)pyrene 9,10-oxide 48-52 glutathione S-transferase theta 1 Homo sapiens 256-261 10463383-0 1999 Haemoglobin adducts of acrylonitrile and ethylene oxide in acrylonitrile workers, dependent on polymorphisms of the glutathione transferases GSTT1 and GSTM1. Acrylonitrile 23-36 glutathione S-transferase theta 1 Homo sapiens 141-146 10463383-0 1999 Haemoglobin adducts of acrylonitrile and ethylene oxide in acrylonitrile workers, dependent on polymorphisms of the glutathione transferases GSTT1 and GSTM1. Ethylene Oxide 41-55 glutathione S-transferase theta 1 Homo sapiens 141-146 10463383-0 1999 Haemoglobin adducts of acrylonitrile and ethylene oxide in acrylonitrile workers, dependent on polymorphisms of the glutathione transferases GSTT1 and GSTM1. Acrylonitrile 59-72 glutathione S-transferase theta 1 Homo sapiens 141-146 10463383-14 1999 The coincidence with known differences in rates of background sister chromatid exchange between GSTT1- and GSTT1 + persons suggests that the lower ethylene oxide (EO) detoxification rate in GSTT1- persons, indicated by elevated blood protein hydroxyethyl adduct levels, leads to an increased genotoxic effect of the physiological EO background. Ethylene Oxide 147-161 glutathione S-transferase theta 1 Homo sapiens 96-101 10463383-14 1999 The coincidence with known differences in rates of background sister chromatid exchange between GSTT1- and GSTT1 + persons suggests that the lower ethylene oxide (EO) detoxification rate in GSTT1- persons, indicated by elevated blood protein hydroxyethyl adduct levels, leads to an increased genotoxic effect of the physiological EO background. Ethylene Oxide 147-161 glutathione S-transferase theta 1 Homo sapiens 107-112 10463383-14 1999 The coincidence with known differences in rates of background sister chromatid exchange between GSTT1- and GSTT1 + persons suggests that the lower ethylene oxide (EO) detoxification rate in GSTT1- persons, indicated by elevated blood protein hydroxyethyl adduct levels, leads to an increased genotoxic effect of the physiological EO background. Ethylene Oxide 147-161 glutathione S-transferase theta 1 Homo sapiens 107-112 10463383-14 1999 The coincidence with known differences in rates of background sister chromatid exchange between GSTT1- and GSTT1 + persons suggests that the lower ethylene oxide (EO) detoxification rate in GSTT1- persons, indicated by elevated blood protein hydroxyethyl adduct levels, leads to an increased genotoxic effect of the physiological EO background. Ethylene Oxide 163-165 glutathione S-transferase theta 1 Homo sapiens 107-112 10463383-14 1999 The coincidence with known differences in rates of background sister chromatid exchange between GSTT1- and GSTT1 + persons suggests that the lower ethylene oxide (EO) detoxification rate in GSTT1- persons, indicated by elevated blood protein hydroxyethyl adduct levels, leads to an increased genotoxic effect of the physiological EO background. Ethylene Oxide 163-165 glutathione S-transferase theta 1 Homo sapiens 107-112 10207629-4 1999 The probability of having CaP was increased in men who had nondeleted (functional) genotypes at GSTT1 (odds ratio, 1.83; 95% confidence interval, 1.19-2.80) but not GSTM1 (odds ratio, 1.07; 95% confidence interval, 0.74-1.55). cap 26-29 glutathione S-transferase theta 1 Homo sapiens 96-101 10350187-2 1999 We found that individuals with genotypes positive for both GST T1 and GST M1 showed the highest prevalence of low WBC [odds ratio (OR) = 4.67, P = 0.046, 95% confidence interval (CI) = 1.02-24.15] when the benzene exposure was high. Benzene 206-213 glutathione S-transferase theta 1 Homo sapiens 59-76 10023062-1 1999 Induction of sister chromatid exchanges (SCEs) by 1,2-epoxy-3-butene (monoepoxybutene, MEB), an epoxide metabolite of 1,3-butadiene, in human whole-blood lymphocyte cultures has previously been observed to depend on the glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) genotype of the blood donor. 3,4-epoxy-1-butene 50-68 glutathione S-transferase theta 1 Homo sapiens 265-270 10023062-1 1999 Induction of sister chromatid exchanges (SCEs) by 1,2-epoxy-3-butene (monoepoxybutene, MEB), an epoxide metabolite of 1,3-butadiene, in human whole-blood lymphocyte cultures has previously been observed to depend on the glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) genotype of the blood donor. 3,4-epoxy-1-butene 70-85 glutathione S-transferase theta 1 Homo sapiens 265-270 9918129-1 1998 Although some blood parameters have been suggested to modulate in-vitro induction of sister chromatid exchanges by 1,2:3,4-diepoxybutane (DEB), a metabolite of 1,3-butadiene, the increased sensitivity has largely been assigned to a homozygous deletion of glutathione S-transferase T1 gene (GSTT1 null genotype). diepoxybutane 115-136 glutathione S-transferase theta 1 Homo sapiens 255-283 9950079-0 1998 Determination of glutathione transferase (GSTT1-1) activities in different tissues based on formation of radioactive metabolites using 35S-glutathione. 35s-glutathione 135-150 glutathione S-transferase theta 1 Homo sapiens 42-49 9950079-1 1998 A new system has been developed to determine enzyme activities of glutathione transferase theta (GSTT1-1) based on radiometric product detection resulting from the enzymic reaction of methyl chloride with 35S-labelled glutathione. Methyl Chloride 184-199 glutathione S-transferase theta 1 Homo sapiens 97-104 9950079-1 1998 A new system has been developed to determine enzyme activities of glutathione transferase theta (GSTT1-1) based on radiometric product detection resulting from the enzymic reaction of methyl chloride with 35S-labelled glutathione. Sulfur-35 205-208 glutathione S-transferase theta 1 Homo sapiens 97-104 9950079-1 1998 A new system has been developed to determine enzyme activities of glutathione transferase theta (GSTT1-1) based on radiometric product detection resulting from the enzymic reaction of methyl chloride with 35S-labelled glutathione. Glutathione 66-77 glutathione S-transferase theta 1 Homo sapiens 97-104 9918129-1 1998 Although some blood parameters have been suggested to modulate in-vitro induction of sister chromatid exchanges by 1,2:3,4-diepoxybutane (DEB), a metabolite of 1,3-butadiene, the increased sensitivity has largely been assigned to a homozygous deletion of glutathione S-transferase T1 gene (GSTT1 null genotype). diepoxybutane 115-136 glutathione S-transferase theta 1 Homo sapiens 290-295 9918129-1 1998 Although some blood parameters have been suggested to modulate in-vitro induction of sister chromatid exchanges by 1,2:3,4-diepoxybutane (DEB), a metabolite of 1,3-butadiene, the increased sensitivity has largely been assigned to a homozygous deletion of glutathione S-transferase T1 gene (GSTT1 null genotype). diepoxybutane 138-141 glutathione S-transferase theta 1 Homo sapiens 255-283 9918129-1 1998 Although some blood parameters have been suggested to modulate in-vitro induction of sister chromatid exchanges by 1,2:3,4-diepoxybutane (DEB), a metabolite of 1,3-butadiene, the increased sensitivity has largely been assigned to a homozygous deletion of glutathione S-transferase T1 gene (GSTT1 null genotype). diepoxybutane 138-141 glutathione S-transferase theta 1 Homo sapiens 290-295 9829702-5 1998 These include the conserved Theta class residues Arg 107, Trp 115, and the conserved GSTT1 subclass residue His 176. Histidine 108-111 glutathione S-transferase theta 1 Homo sapiens 85-90 9794803-13 1998 It is concluded that the induction of GSTT1 by dietary components and its localization within cells are important factors that should be considered when assessing the risk dihaloalkanes pose to human health. dihaloalkanes 172-185 glutathione S-transferase theta 1 Homo sapiens 38-43 9285043-0 1997 Influence of polymorphisms of GSTM1 and GSTT1 for risk of renal cell cancer in workers with long-term high occupational exposure to trichloroethene. Trichloroethylene 132-147 glutathione S-transferase theta 1 Homo sapiens 40-45 9851677-0 1998 Species differences in the glutathione transferase GSTT1-1 activity towards the model substrates methyl chloride and dichloromethane in liver and kidney. methyl chloride and dichloromethane 97-132 glutathione S-transferase theta 1 Homo sapiens 51-58 9851677-1 1998 Glutathione transferase (GST) GSTT1-1 is involved in the biotransformation of several chemicals widely used in industry, such as butadiene and dichloro methane DCM. 1,3-butadiene 129-138 glutathione S-transferase theta 1 Homo sapiens 30-37 9851677-1 1998 Glutathione transferase (GST) GSTT1-1 is involved in the biotransformation of several chemicals widely used in industry, such as butadiene and dichloro methane DCM. dichloro methane dcm 143-163 glutathione S-transferase theta 1 Homo sapiens 30-37 9851677-2 1998 The polymorphic hGSTT1-1 may well play a role in the development of kidney tumours after high and long-term occupational exposure against trichloroethylene. Trichloroethylene 138-155 glutathione S-transferase theta 1 Homo sapiens 16-24 9851677-5 1998 In addition GSTT1-1 activity towards MC and DCM was determined in human erythrocytes. Methyl Chloride 37-39 glutathione S-transferase theta 1 Homo sapiens 12-19 9851677-5 1998 In addition GSTT1-1 activity towards MC and DCM was determined in human erythrocytes. dcm 44-47 glutathione S-transferase theta 1 Homo sapiens 12-19 9851677-9 1998 GSTT1-1 activity towards MC was two to seven-times higher in liver cytosol than in kidney cytosol. Methyl Chloride 25-27 glutathione S-transferase theta 1 Homo sapiens 0-7 9438048-4 1998 We also sought to investigate whether the glutathione S-transferase class theta gene (GSTT1) affects individual susceptibility to cytogenetic damage induced by in vivo exposure to benzene or in vitro exposure to diepoxybutane. Benzene 180-187 glutathione S-transferase theta 1 Homo sapiens 86-91 9438048-4 1998 We also sought to investigate whether the glutathione S-transferase class theta gene (GSTT1) affects individual susceptibility to cytogenetic damage induced by in vivo exposure to benzene or in vitro exposure to diepoxybutane. diepoxybutane 212-225 glutathione S-transferase theta 1 Homo sapiens 86-91 9438048-9 1998 A significant benzene-GSTT1 interaction was found in nonsmokers (P < 0.05). Benzene 14-21 glutathione S-transferase theta 1 Homo sapiens 22-27 9438048-10 1998 Our study suggests that GSTT1 is an important determinant of heterogeneity in individual susceptibility to chromosomal damage associated with exposure to benzene. Benzene 154-161 glutathione S-transferase theta 1 Homo sapiens 24-29 9498293-1 1998 The influence of glutathione S-transferase T1 (GSTT1) genotype on the genotoxicity of 1,2-epoxy-3-butene (MEB), a metabolite of 1,3-butadiene, was assessed by the analysis of sister chromatid exchanges (SCEs) in 72-h human whole-blood lymphocyte cultures. 3,4-epoxy-1-butene 86-104 glutathione S-transferase theta 1 Homo sapiens 17-45 9498293-1 1998 The influence of glutathione S-transferase T1 (GSTT1) genotype on the genotoxicity of 1,2-epoxy-3-butene (MEB), a metabolite of 1,3-butadiene, was assessed by the analysis of sister chromatid exchanges (SCEs) in 72-h human whole-blood lymphocyte cultures. 3,4-epoxy-1-butene 86-104 glutathione S-transferase theta 1 Homo sapiens 47-52 9539016-7 1998 From all biomarkers the effect of GSTM1 and N-acetyl transferase 2 was seen in coke oven workers on mutagenicity of urine and of glutathione S-transferase T1 on the chromosomal aberrations in subjects from 1,3-butadiene monomer production units. 1,3-butadiene 206-219 glutathione S-transferase theta 1 Homo sapiens 129-157 9654239-0 1998 Influence of GSTT1 genotype on sister chromatid exchange induction by styrene-7,8-oxide in cultured human lymphocytes. styrene oxide 70-87 glutathione S-transferase theta 1 Homo sapiens 13-18 9654239-8 1998 Although glutathione conjugation is considered a minor metabolic pathway for SO in vivo, the high GSTT1 activity in erythrocytes may be important locally and might affect the level of genotoxic damage observed in peripheral lymphocytes of styrene-exposed reinforced plastics workers. Styrene 239-246 glutathione S-transferase theta 1 Homo sapiens 98-103 9654239-9 1998 The GSTT1 polymorphism could also influence the urinary excretion of SO-specific mercapturic acids. Acetylcysteine 81-98 glutathione S-transferase theta 1 Homo sapiens 4-9 10026993-11 1998 In individuals with the GSTT1-0 genotype more chromosomal aberrations and sister chromatid exchanges (SCEs) were observed after exposure to 1,3-butadiene or various haloalkanes or haloalkenes. 1,3-butadiene 140-153 glutathione S-transferase theta 1 Homo sapiens 24-29 10026993-11 1998 In individuals with the GSTT1-0 genotype more chromosomal aberrations and sister chromatid exchanges (SCEs) were observed after exposure to 1,3-butadiene or various haloalkanes or haloalkenes. haloalkanes 165-176 glutathione S-transferase theta 1 Homo sapiens 24-29 10026993-11 1998 In individuals with the GSTT1-0 genotype more chromosomal aberrations and sister chromatid exchanges (SCEs) were observed after exposure to 1,3-butadiene or various haloalkanes or haloalkenes. haloalkenes 180-191 glutathione S-transferase theta 1 Homo sapiens 24-29 9379921-0 1997 GSTT1-dependent induction of centromere-negative and -positive micronuclei by 1,2:3,4-diepoxybutane in cultured human lymphocytes. diepoxybutane 78-99 glutathione S-transferase theta 1 Homo sapiens 0-5 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. diepoxybutane 95-116 glutathione S-transferase theta 1 Homo sapiens 16-44 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. diepoxybutane 95-116 glutathione S-transferase theta 1 Homo sapiens 51-56 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. diepoxybutane 118-121 glutathione S-transferase theta 1 Homo sapiens 16-44 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. diepoxybutane 118-121 glutathione S-transferase theta 1 Homo sapiens 51-56 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. Epoxy Compounds 127-134 glutathione S-transferase theta 1 Homo sapiens 16-44 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. Epoxy Compounds 127-134 glutathione S-transferase theta 1 Homo sapiens 51-56 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. 1,3-butadiene 149-162 glutathione S-transferase theta 1 Homo sapiens 16-44 9379921-1 1997 The role of the glutathione S-transferase T1 gene (GSTT1) in determining genotoxic response to 1,2:3,4-diepoxybutane (DEB), an epoxide metabolite of 1,3-butadiene, was studied by analysis of micronuclei (MN) in cultured human lymphocytes using the cytokinesis block method. 1,3-butadiene 149-162 glutathione S-transferase theta 1 Homo sapiens 51-56 9241666-0 1997 Glutathione S-transferase theta genetic polymorphism might influence tacrine hepatotoxicity in Alzheimer"s patients. Tacrine 69-76 glutathione S-transferase theta 1 Homo sapiens 0-31 9826084-17 1998 CONCLUSIONS: This result is in accordance with the finding that ethylene oxide is a substrate for GSTT1 but not for GSTM1. Ethylene Oxide 64-78 glutathione S-transferase theta 1 Homo sapiens 98-103 9826084-18 1998 In addition, this study demonstrates a clear influence of genetically determined GSTT1 status on biological effects, e.g., protein adduct formation after non-occupational ethylene oxide exposure. Ethylene Oxide 171-185 glutathione S-transferase theta 1 Homo sapiens 81-86 10022746-1 1998 Glutathione S-transferase GSTM1, GSTM3 and GSTT1 and cytochrome P450 CYP2D6, CYP1A1 and CYP2E1 loci are susceptibility candidates for cancers of the upper aerodigestive tract because putatively protective and risk genotypes have been identified from studies in other diseases associated with alcohol and tobacco consumption. Alcohols 292-299 glutathione S-transferase theta 1 Homo sapiens 43-48 9511178-1 1998 Deficiencies of the glutathione transferase isoenzymes GSTM1-1 and GSTT1-1 have been shown to be risk modifiers in a number of different cancers but there have been no similar studies with GSTP1-1, the only member of the Pi class of glutathione S-transferases expressed in humans. Glutathione 20-31 glutathione S-transferase theta 1 Homo sapiens 67-74 9434735-1 1997 Recombinant human theta class glutathione transferase T1-1 has been heterologously expressed in Escherichia coli and a simple purification method involving immobilized ferric ion affinity chromatography and Orange A dye chromatography is described. Ferric enterobactin ion 168-174 glutathione S-transferase theta 1 Homo sapiens 30-58 9434735-3 1997 In addition to 1,2-epoxy-3-(4-nitrophenoxy)propane, the substrate used previously to monitor the enzyme, human glutathione transferase T1-1 has activity with the naturally occurring phenethylisothiocyanate and also displays glutathione peroxidase activity with cumene hydroperoxide. 1,2-epoxy-3-(p-nitrophenoxy)propane 15-50 glutathione S-transferase theta 1 Homo sapiens 111-139 9434735-3 1997 In addition to 1,2-epoxy-3-(4-nitrophenoxy)propane, the substrate used previously to monitor the enzyme, human glutathione transferase T1-1 has activity with the naturally occurring phenethylisothiocyanate and also displays glutathione peroxidase activity with cumene hydroperoxide. phenethyl isothiocyanate 182-205 glutathione S-transferase theta 1 Homo sapiens 111-139 9434735-3 1997 In addition to 1,2-epoxy-3-(4-nitrophenoxy)propane, the substrate used previously to monitor the enzyme, human glutathione transferase T1-1 has activity with the naturally occurring phenethylisothiocyanate and also displays glutathione peroxidase activity with cumene hydroperoxide. cumene hydroperoxide 261-281 glutathione S-transferase theta 1 Homo sapiens 111-139 9307035-2 1997 The cDNA encoding human glutathione S-transferase (GST) T1 has been expressed as two recombinant forms in Escherichia coli that could be purified by affinity chromatography on either IgG-Sepharose or nickel-agarose; one form of the transferase was synthesized from the pALP 1 expression vector as a Staphylococcus aureus protein A fusion, whereas the other form was synthesized from the pET-20b expression vector as a C-terminal polyhistidine-tagged recombinant. Sepharose 187-196 glutathione S-transferase theta 1 Homo sapiens 24-58 9307035-2 1997 The cDNA encoding human glutathione S-transferase (GST) T1 has been expressed as two recombinant forms in Escherichia coli that could be purified by affinity chromatography on either IgG-Sepharose or nickel-agarose; one form of the transferase was synthesized from the pALP 1 expression vector as a Staphylococcus aureus protein A fusion, whereas the other form was synthesized from the pET-20b expression vector as a C-terminal polyhistidine-tagged recombinant. Nickel 200-206 glutathione S-transferase theta 1 Homo sapiens 24-58 9307035-2 1997 The cDNA encoding human glutathione S-transferase (GST) T1 has been expressed as two recombinant forms in Escherichia coli that could be purified by affinity chromatography on either IgG-Sepharose or nickel-agarose; one form of the transferase was synthesized from the pALP 1 expression vector as a Staphylococcus aureus protein A fusion, whereas the other form was synthesized from the pET-20b expression vector as a C-terminal polyhistidine-tagged recombinant. Sepharose 207-214 glutathione S-transferase theta 1 Homo sapiens 24-58 9307035-2 1997 The cDNA encoding human glutathione S-transferase (GST) T1 has been expressed as two recombinant forms in Escherichia coli that could be purified by affinity chromatography on either IgG-Sepharose or nickel-agarose; one form of the transferase was synthesized from the pALP 1 expression vector as a Staphylococcus aureus protein A fusion, whereas the other form was synthesized from the pET-20b expression vector as a C-terminal polyhistidine-tagged recombinant. polyhistidine 429-442 glutathione S-transferase theta 1 Homo sapiens 24-58 8671741-0 1996 Influence of erythrocyte glutathione S-transferase T1 on sister chromatid exchanges induced by diepoxybutane in cultured human lymphocytes. diepoxybutane 95-108 glutathione S-transferase theta 1 Homo sapiens 25-53 8980697-0 1996 Induction of sister chromatid exchange by 3,4-expoxybutane-1,2-diol in cultured human lymphocytes of different GSTT1 and GSTM1 genotypes. 3,4-expoxybutane-1,2-diol 42-67 glutathione S-transferase theta 1 Homo sapiens 111-116 8980697-1 1996 The induction of sister chromatid exchanges (SCEs) by a 48-h treatment with 3,4-epoxybutane-1,2-diol (EBD), a metabolite of 1,3-butadiene, was studied in whole-blood lymphocyte cultures of 22 human donors with known genotypes of two polymorphic glutathione S-transferases (GSTs), GSTT1 and GSTM1. 3,4-epoxybutane-1,2-diol 76-100 glutathione S-transferase theta 1 Homo sapiens 280-285 8799324-1 1996 A high activity glutathione S-transferase T1-1 (GSTT1-1) towards dichloromethane was isolated from human liver cytosol and purified to homogenity in 18.5% yield with a purification factor of 4400-fold. Glutathione 16-27 glutathione S-transferase theta 1 Homo sapiens 48-55 8799324-1 1996 A high activity glutathione S-transferase T1-1 (GSTT1-1) towards dichloromethane was isolated from human liver cytosol and purified to homogenity in 18.5% yield with a purification factor of 4400-fold. Methylene Chloride 65-80 glutathione S-transferase theta 1 Homo sapiens 48-55 8799324-3 1996 The purified GSTT1-1-s were homo-dimeric enzymes with a subunit M1 value 25,300 and pI 6 64, as confirmed by SDS-PAGE, IEF and Western blot analysis. Sodium Dodecyl Sulfate 109-112 glutathione S-transferase theta 1 Homo sapiens 13-20 9064289-1 1996 The recently discovered human class theta glutathione S-transferase T1-1 (GSTT1-1) is responsible for the GSH-dependent detoxification of naturally occurring monohalomethanes. Glutathione 42-53 glutathione S-transferase theta 1 Homo sapiens 74-81 8569364-0 1996 Increased risk for myelodysplastic syndromes in individuals with glutathione transferase theta 1 (GSTT1) gene defect. Glutathione 65-76 glutathione S-transferase theta 1 Homo sapiens 98-103 8704864-2 1996 To examine the influence of the polymorphism of glutathione-S-transferase theta (GSTT1) on the neurotoxicity of methyl bromide. methyl bromide 112-126 glutathione S-transferase theta 1 Homo sapiens 48-79 8704864-2 1996 To examine the influence of the polymorphism of glutathione-S-transferase theta (GSTT1) on the neurotoxicity of methyl bromide. methyl bromide 112-126 glutathione S-transferase theta 1 Homo sapiens 81-86 9064289-1 1996 The recently discovered human class theta glutathione S-transferase T1-1 (GSTT1-1) is responsible for the GSH-dependent detoxification of naturally occurring monohalomethanes. Glutathione 106-109 glutathione S-transferase theta 1 Homo sapiens 74-81 9064289-1 1996 The recently discovered human class theta glutathione S-transferase T1-1 (GSTT1-1) is responsible for the GSH-dependent detoxification of naturally occurring monohalomethanes. monohalomethanes 158-174 glutathione S-transferase theta 1 Homo sapiens 74-81 8524342-6 1995 Recent work has further demonstrated that cultured lymphocytes from the approximately 20% of the Caucasian population that lack the glutathione S-transferase class theta gene (GSTT1) are relatively sensitive to the induction of cytogenetic damage by butadiene metabolites. Glutathione 132-143 glutathione S-transferase theta 1 Homo sapiens 176-181 8852702-5 1996 The GSTT1-1 phenotype was determined by measuring the erythrocyte conjugating activity towards methyl chloride using a gas chromatographic assay. Methyl Chloride 95-110 glutathione S-transferase theta 1 Homo sapiens 4-11 8831906-3 1996 Although the presence or absence of the enzyme activity in human red blood cells is parallel with the polymorphism of the human GST T1 gene, the new GST theta in red blood cells may differ from the known GST T1-1 enzyme from other tissues in terms of substrate specificity, since established GST T1-1 substrates [1,2-epoxy-3-(p-nitro-phenoxy)propane and p-nitro-benzyl chloride] are not metabolized. 1,2-epoxy-3-(p-nitrophenoxy)propane 313-349 glutathione S-transferase theta 1 Homo sapiens 128-134 8831906-3 1996 Although the presence or absence of the enzyme activity in human red blood cells is parallel with the polymorphism of the human GST T1 gene, the new GST theta in red blood cells may differ from the known GST T1-1 enzyme from other tissues in terms of substrate specificity, since established GST T1-1 substrates [1,2-epoxy-3-(p-nitro-phenoxy)propane and p-nitro-benzyl chloride] are not metabolized. 4-nitrobenzyl chloride 354-377 glutathione S-transferase theta 1 Homo sapiens 128-134 8565128-5 1996 Mutagenicity of EDB, BDE, CH2Br2, epibromohydrin and 1,3-dichloroacetone was higher in the S.typhimurium TA1535 expressing GSTT1-1 than in the control strain. Ethylene Dibromide 16-19 glutathione S-transferase theta 1 Homo sapiens 123-128 8565128-5 1996 Mutagenicity of EDB, BDE, CH2Br2, epibromohydrin and 1,3-dichloroacetone was higher in the S.typhimurium TA1535 expressing GSTT1-1 than in the control strain. methylene bromide 26-32 glutathione S-transferase theta 1 Homo sapiens 123-128 8565128-5 1996 Mutagenicity of EDB, BDE, CH2Br2, epibromohydrin and 1,3-dichloroacetone was higher in the S.typhimurium TA1535 expressing GSTT1-1 than in the control strain. Epibromohydrin 34-48 glutathione S-transferase theta 1 Homo sapiens 123-128 8565128-5 1996 Mutagenicity of EDB, BDE, CH2Br2, epibromohydrin and 1,3-dichloroacetone was higher in the S.typhimurium TA1535 expressing GSTT1-1 than in the control strain. 1,3-dichloroacetone 53-72 glutathione S-transferase theta 1 Homo sapiens 123-128 8524342-6 1995 Recent work has further demonstrated that cultured lymphocytes from the approximately 20% of the Caucasian population that lack the glutathione S-transferase class theta gene (GSTT1) are relatively sensitive to the induction of cytogenetic damage by butadiene metabolites. 1,3-butadiene 250-259 glutathione S-transferase theta 1 Homo sapiens 176-181 33804102-3 2021 METHODS: We conducted a multivariate analysis involving 177 subjects to determine the association between paraben concentrations and birth outcomes in mothers with GST mu 1 (GSTM1) and GST theta 1 (GSTT1) polymorphisms from 2017 to 2019. Parabens 106-113 glutathione S-transferase theta 1 Homo sapiens 185-196 7606200-10 1995 Individuals who carry a homozygous deletion of the GSTT1 gene may be at increased risk for genotoxic damage from environmental or occupational 1,3-butadiene exposures. 1,3-butadiene 143-156 glutathione S-transferase theta 1 Homo sapiens 51-56 8526747-1 1995 Polymorphism of glutathione S-transferase theta (GSTT1) modulates the toxicity of halogenated alkanes and epoxides in humans. Alkanes 94-101 glutathione S-transferase theta 1 Homo sapiens 16-47 8526747-1 1995 Polymorphism of glutathione S-transferase theta (GSTT1) modulates the toxicity of halogenated alkanes and epoxides in humans. Alkanes 94-101 glutathione S-transferase theta 1 Homo sapiens 49-54 8526747-1 1995 Polymorphism of glutathione S-transferase theta (GSTT1) modulates the toxicity of halogenated alkanes and epoxides in humans. Epoxy Compounds 106-114 glutathione S-transferase theta 1 Homo sapiens 16-47 8526747-1 1995 Polymorphism of glutathione S-transferase theta (GSTT1) modulates the toxicity of halogenated alkanes and epoxides in humans. Epoxy Compounds 106-114 glutathione S-transferase theta 1 Homo sapiens 49-54 8526747-9 1995 Since ethylene oxide is a proven substrate of GSTT1, the detoxification of this epoxide arising from endogenous ethylene may modulate SCE background rates. Ethylene Oxide 6-20 glutathione S-transferase theta 1 Homo sapiens 46-51 8526747-9 1995 Since ethylene oxide is a proven substrate of GSTT1, the detoxification of this epoxide arising from endogenous ethylene may modulate SCE background rates. Epoxy Compounds 80-87 glutathione S-transferase theta 1 Homo sapiens 46-51 8526747-9 1995 Since ethylene oxide is a proven substrate of GSTT1, the detoxification of this epoxide arising from endogenous ethylene may modulate SCE background rates. ethylene 6-14 glutathione S-transferase theta 1 Homo sapiens 46-51 8198545-6 1994 The GSTT1+ phenotype can catalyse the glutathione conjugation of dichloromethane, a metabolic pathway which has been shown to be mutagenic in Salmonella typhimurium mutagenicity tester strains and is believed to be responsible for carcinogenicity of dichloromethane in the mouse. Glutathione 38-49 glutathione S-transferase theta 1 Homo sapiens 4-9 8198545-6 1994 The GSTT1+ phenotype can catalyse the glutathione conjugation of dichloromethane, a metabolic pathway which has been shown to be mutagenic in Salmonella typhimurium mutagenicity tester strains and is believed to be responsible for carcinogenicity of dichloromethane in the mouse. Methylene Chloride 65-80 glutathione S-transferase theta 1 Homo sapiens 4-9 8198545-6 1994 The GSTT1+ phenotype can catalyse the glutathione conjugation of dichloromethane, a metabolic pathway which has been shown to be mutagenic in Salmonella typhimurium mutagenicity tester strains and is believed to be responsible for carcinogenicity of dichloromethane in the mouse. Methylene Chloride 250-265 glutathione S-transferase theta 1 Homo sapiens 4-9 8198545-8 1994 Characterization of the GSTT1 polymorphism will thus enable a more accurate assessment of human health risk from synthetic halomethanes and other industrial chemicals. halomethanes 123-135 glutathione S-transferase theta 1 Homo sapiens 24-29 33804102-8 2021 We found positive relationships of maternal exposure to methyl parabens with birth weight in both mothers with GSTM1 and GSTT1-null genotypes. methylparaben 56-71 glutathione S-transferase theta 1 Homo sapiens 121-126 7485742-2 1995 The absence of glutathione S-transferases mu (GSTM1) and theta (GSTT1) results in decreased detoxification of carcinogens, for example, chemicals in cigarette smoke. Glutathione 15-26 glutathione S-transferase theta 1 Homo sapiens 64-69 7788840-0 1995 Role of GSTT1 and GSTM1 genotypes in determining individual sensitivity to sister chromatid exchange induction by diepoxybutane in cultured human lymphocytes. diepoxybutane 114-127 glutathione S-transferase theta 1 Homo sapiens 8-13 7788840-8 1995 A significant (P < 0.05) negative correlation (r = -0.65 at 5 microM, r = -0.56 at 2 microM) was obtained in the GSTT1 positive donors between DEB-induced individual SCE frequency and RBC GSTT1 activity, measured by formaldehyde formation from dichloromethane; the GSTT1 null individuals showed no GSTT1 activity. Formaldehyde 219-231 glutathione S-transferase theta 1 Homo sapiens 116-121 7788840-8 1995 A significant (P < 0.05) negative correlation (r = -0.65 at 5 microM, r = -0.56 at 2 microM) was obtained in the GSTT1 positive donors between DEB-induced individual SCE frequency and RBC GSTT1 activity, measured by formaldehyde formation from dichloromethane; the GSTT1 null individuals showed no GSTT1 activity. Methylene Chloride 247-262 glutathione S-transferase theta 1 Homo sapiens 116-121 33804102-4 2021 Furthermore, we determined the interactive effect between paraben levels and GSTM1/GSTT1 polymorphisms using regression analysis, in addition to a generalized linear model after stratifying GSTM1/GSTT1 genotype into three categories. Parabens 58-65 glutathione S-transferase theta 1 Homo sapiens 83-88 34453679-7 2022 The metal-gene interaction showed positive correlation between GSTT1 null genotype and Pb and Cd levels (beta = 0.11; p = 0.02 and beta = 0.10; p = 0.01, respectively). Metals 4-9 glutathione S-transferase theta 1 Homo sapiens 63-68 34499222-1 2022 PURPOSE: This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. Glutathione 98-109 glutathione S-transferase theta 1 Homo sapiens 135-140 34499222-1 2022 PURPOSE: This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. Busulfan 225-233 glutathione S-transferase theta 1 Homo sapiens 135-140 34499222-1 2022 PURPOSE: This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. Busulfan 235-237 glutathione S-transferase theta 1 Homo sapiens 135-140 34499222-1 2022 PURPOSE: This study aimed to retrospectively evaluate the genetic association of null variants of glutathione S-transferases GSTM1 and GSTT1 with relapse incidence in children with hematological malignancies (HMs) undergoing busulfan (BU)- containing allogeneic hematopoietic stem cell transplantation (HSCT) and to assess the impact of these variants on BU-induced cytotoxicity on the immortalized lymphoblastoid cell lines (LCLs) and tumor THP1 GST gene-edited cell models. Busulfan 355-357 glutathione S-transferase theta 1 Homo sapiens 135-140 34499222-6 2022 BU-induced cell death preferentially in THP1GSTM1(non-null) and LCLsGSTM1(non-null) as shown by decreased viability, increased necrosis and levels of the oxidized form of glutathione compared to null cells, while GSTT1 non-null cells showed increased baseline proliferation. Busulfan 0-2 glutathione S-transferase theta 1 Homo sapiens 213-218 34453679-7 2022 The metal-gene interaction showed positive correlation between GSTT1 null genotype and Pb and Cd levels (beta = 0.11; p = 0.02 and beta = 0.10; p = 0.01, respectively). Lead 87-89 glutathione S-transferase theta 1 Homo sapiens 63-68 34453679-7 2022 The metal-gene interaction showed positive correlation between GSTT1 null genotype and Pb and Cd levels (beta = 0.11; p = 0.02 and beta = 0.10; p = 0.01, respectively). Cadmium 94-96 glutathione S-transferase theta 1 Homo sapiens 63-68 34453679-10 2022 Further, the combined effect of GSTP1 Ile/Val with GSTT1 null genotype was more pronounced and showed an increased risk of susceptibility to Pb toxicity (OR = 11.7; 95% CI: 1.36-102.1, p = 0.02). Lead 141-143 glutathione S-transferase theta 1 Homo sapiens 51-56 34453679-11 2022 In summary, this study suggests that GSTT1 null and GSTP1 Ile/Val genotypes are the main genetic factors, and individual and specific combinations of GSTP1 Ile/Val with GSTM1 and GSTT1 GST polymorphisms are associated with susceptibility to Pb toxicity. Lead 241-243 glutathione S-transferase theta 1 Homo sapiens 37-42 34453679-11 2022 In summary, this study suggests that GSTT1 null and GSTP1 Ile/Val genotypes are the main genetic factors, and individual and specific combinations of GSTP1 Ile/Val with GSTM1 and GSTT1 GST polymorphisms are associated with susceptibility to Pb toxicity. Lead 241-243 glutathione S-transferase theta 1 Homo sapiens 179-184 34766662-9 2022 GMDR revealed a significant interaction between GSTT1 and LL55 genotype. gmdr 0-4 glutathione S-transferase theta 1 Homo sapiens 48-53 34992428-1 2021 Purpose: Glutathione S-transferases (GSTT1 and GSTM1) are instrumental in detoxification process of activated carcinogens. Glutathione 9-20 glutathione S-transferase theta 1 Homo sapiens 37-42 34965566-7 2021 It wasfound that in children with BA the tendency to frequency of the deletion variant of the GSTT1 and GSTM1 genes incomparison with children without bronchial and pulmonary pathology was increased. Barium 34-36 glutathione S-transferase theta 1 Homo sapiens 94-99 34965566-14 2021 One of the leading mechanisms, due to which there is a realization of hereditary predisposition tobronchial asthma in children living under constant intake of radionuclides with a long half-life, is the polymorphismof certain glutathione-S-transferase genes, namely, GSTT1, GSTM1 and A313G gene deletion polymorphism and GSTP1gene polymorphism. Radioisotopes 159-172 glutathione S-transferase theta 1 Homo sapiens 267-272 32892263-0 2021 Interaction of Blood Manganese Concentrations with GSTT1 in Relation to Autism Spectrum Disorder in Jamaican Children. Manganese 21-30 glutathione S-transferase theta 1 Homo sapiens 51-56 34535163-0 2021 Up-regulation of GSTT1 in serous ovarian cancer associated with resistance to TAXOL / carboplatin. Paclitaxel 78-83 glutathione S-transferase theta 1 Homo sapiens 17-22 34535163-0 2021 Up-regulation of GSTT1 in serous ovarian cancer associated with resistance to TAXOL / carboplatin. Carboplatin 86-97 glutathione S-transferase theta 1 Homo sapiens 17-22 34535163-6 2021 Meanwhile, GSTT1 expression was also significantly upregulated in the SOC patient tissues after taxol treatment, indicating this upregulation was physiologically relevant to chemotherapy. Paclitaxel 96-101 glutathione S-transferase theta 1 Homo sapiens 11-16 34437496-0 2021 Formaldehyde in Hospitals Induces Oxidative Stress: The Role of GSTT1 and GSTM1 Polymorphisms. Formaldehyde 0-12 glutathione S-transferase theta 1 Homo sapiens 64-69 34437496-8 2021 In the formalin-employers group the MDA level was significantly higher in GSTT1 Null (p = 0.038), GSTM1 Null (p = 0.031), and CYP1A1 exon 7 mutation carrier (p = 0.008) workers, compared to the wild type subjects. Formaldehyde 7-15 glutathione S-transferase theta 1 Homo sapiens 74-79 34295994-9 2021 Furthermore, some polymorphisms on NQO1, GSTT1, GSTM1, MPO, and CYP2E1, among other genes, showed a statistically significant relationship with an increased risk of developing at least one of these effects on benzene-exposed workers. Benzene 209-216 glutathione S-transferase theta 1 Homo sapiens 41-46 34437496-8 2021 In the formalin-employers group the MDA level was significantly higher in GSTT1 Null (p = 0.038), GSTM1 Null (p = 0.031), and CYP1A1 exon 7 mutation carrier (p = 0.008) workers, compared to the wild type subjects. Malondialdehyde 36-39 glutathione S-transferase theta 1 Homo sapiens 74-79 34754473-3 2021 We sought to examine the association between prenatal PAH exposure and infantile allergic diseases in 6-month-old infants, and how maternal glutathione S-transferase M1 (GSTM1) or T1 (GSTT1) polymorphism affects the association between prenatal PAH exposure and allergic diseases in the Mothers and Children"s Environmental Health (MOCEH) study. Polycyclic Aromatic Hydrocarbons 245-248 glutathione S-transferase theta 1 Homo sapiens 184-189 34754473-9 2021 The increased risk of infantile allergic diseases associated with urinary 1-OHP during the early period of pregnancy was limited to the maternal GSTT1 null type (OR: 2.69; 95% CI: 1.17, 6.21, by one log-transformed unit of 1-OHP mug/g creatinine); however, the Relative Excess Risk due to Interaction was not statistically significant. Oxaliplatin 74-79 glutathione S-transferase theta 1 Homo sapiens 145-150 34754473-9 2021 The increased risk of infantile allergic diseases associated with urinary 1-OHP during the early period of pregnancy was limited to the maternal GSTT1 null type (OR: 2.69; 95% CI: 1.17, 6.21, by one log-transformed unit of 1-OHP mug/g creatinine); however, the Relative Excess Risk due to Interaction was not statistically significant. Oxaliplatin 223-228 glutathione S-transferase theta 1 Homo sapiens 145-150 34754473-9 2021 The increased risk of infantile allergic diseases associated with urinary 1-OHP during the early period of pregnancy was limited to the maternal GSTT1 null type (OR: 2.69; 95% CI: 1.17, 6.21, by one log-transformed unit of 1-OHP mug/g creatinine); however, the Relative Excess Risk due to Interaction was not statistically significant. Creatinine 235-245 glutathione S-transferase theta 1 Homo sapiens 145-150 34754473-10 2021 Conclusions: The present study found that infantile allergic diseases could be affected by intrauterine PAH exposure, particularly in the early prenatal period and the risk was limited to the maternal GSTT1 null type. Polycyclic Aromatic Hydrocarbons 104-107 glutathione S-transferase theta 1 Homo sapiens 201-206 35614607-1 2022 OBJECTIVE: To assess the association of single nucleotide polymorphisms in fatty acid binding protein-2 (rs1799883) and glutathione S-transferase pi (rs1695) genes with presence/absence of glutathione S-transferase mu and glutathione S-transferase theta genes in type 2 diabetes. Glutathione 120-131 glutathione S-transferase theta 1 Homo sapiens 222-253 35621716-4 2022 Deletion polymorphisms of GSTM1 and GSTT1, members of the GST family, are common in humans and are presumed to be associated with cisplatin-induced hearing impairment. Cisplatin 130-139 glutathione S-transferase theta 1 Homo sapiens 36-41 35621716-5 2022 However, the specific roles of GSTM1 and GSTT1 in cisplatin ototoxicity are not completely clear. Cisplatin 50-59 glutathione S-transferase theta 1 Homo sapiens 41-46 35605733-2 2022 We aimed to evaluate the associations between serum concentrations of 18 OCPs and semen quality among 387 Chinese men, and further to examine the modifying effects by genetic polymorphisms in cytochrome P450 (CYP2E1) and glutathione S-transferase (GSTT1). ocps 73-77 glutathione S-transferase theta 1 Homo sapiens 248-253 35605733-10 2022 Our results suggested that CYP2E1 and GSTT1 polymorphisms may modify the effects of OCP exposures on semen quality. 5,10,15,20-tetra(3,5-(nido-carboranylmethyl)phenyl)porphyrin 84-87 glutathione S-transferase theta 1 Homo sapiens 38-43 35527244-9 2022 Whereas, homozygous GSTP1 and GSTT1 null genotype is significantly higher only among nilotinib non-responders. nilotinib 85-94 glutathione S-transferase theta 1 Homo sapiens 30-35 35426585-5 2022 Genetic variants in MTR, PCYT1A, ASS1, SLC 25A13, GSTM1, GSTT1, SUMO1 BHMT1, and BHMT2 are being reported to be linked with CL/P risk. Phosphorus 127-128 glutathione S-transferase theta 1 Homo sapiens 57-62 35313604-1 2022 Purpose: Glutathione S-transferases (GSTT1 and GSTM1) detoxify various endogenous and exogenous compounds and provide cytoprotective role against reactive species. Glutathione 9-20 glutathione S-transferase theta 1 Homo sapiens 37-42 35186174-0 2022 Association of GSTM1 and GSTT1 Null Genotypes with Toluene Diisocyanate-Induced Asthma. Toluene 2,4-Diisocyanate 51-71 glutathione S-transferase theta 1 Homo sapiens 25-30 32772315-0 2021 GSTM1 and GSTT1 Null Genotype Polymorphisms and Susceptibility to Arsenic Poisoning: a Meta-analysis. Arsenic 66-73 glutathione S-transferase theta 1 Homo sapiens 10-15 33544514-8 2021 The interaction between GSTT1 null polymorphism and asbestos exposure decreased the MM risk (OR = 0.17; 95% CI = 0.03-0.85; p = 0.031). Asbestos 52-60 glutathione S-transferase theta 1 Homo sapiens 24-29 33871746-4 2021 The study aimed to investigate metal concentrations in dust, particulate matter and urine of exposed workers and correlate with oxidative stress and glutathione S-transferases genotypes (GSTM1 and GSTT1) that play a role in detoxification of metals in humans. Metals 31-36 glutathione S-transferase theta 1 Homo sapiens 197-202 33871746-4 2021 The study aimed to investigate metal concentrations in dust, particulate matter and urine of exposed workers and correlate with oxidative stress and glutathione S-transferases genotypes (GSTM1 and GSTT1) that play a role in detoxification of metals in humans. Glutathione 149-160 glutathione S-transferase theta 1 Homo sapiens 197-202 33443393-0 2021 The Relationship Between GSTT1, GSTM1, GSTO1, GSTP1 and MTHFR Gene Polymorphisms and DNA Damage of BRCA1 and BRCA2 Genes in Arsenic-Exposed Workers. Arsenic 124-131 glutathione S-transferase theta 1 Homo sapiens 25-30 33709282-3 2021 This study reports, the influence of GSTP1*B and GSTT1/GSTM1null polymorphisms in response to imatinib in CML patients in a Brazilian population. Imatinib Mesylate 94-102 glutathione S-transferase theta 1 Homo sapiens 49-54 33443393-5 2021 CONCLUSIONS: Our findings suggest that the DNA damage levels of BRCA1 and BRCA2 genes may modulate by genetic variations of GSTT1 and GSTO1 when individuals are exposed to carcinogens, such as arsenic. Arsenic 193-200 glutathione S-transferase theta 1 Homo sapiens 124-129 33745084-20 2021 We believed the deletion GSTT1null/GSTM1null may contribute to the increased of the oxidative stress in SLE patients while carriers of the mutant SOD2 47CT/TT allele may have greater oxidative stress due to structural change in the protein and decreased H2O2 production. Hydrogen Peroxide 254-258 glutathione S-transferase theta 1 Homo sapiens 25-30 33600611-2 2022 Glutathione S-transferase (GSTT1) is involved in activation of detoxification reactions and catalysis of chemicals conjugation with glutathione. Glutathione 0-11 glutathione S-transferase theta 1 Homo sapiens 27-32 33600611-2 2022 Glutathione S-transferase (GSTT1) is involved in activation of detoxification reactions and catalysis of chemicals conjugation with glutathione. Glutathione 132-143 glutathione S-transferase theta 1 Homo sapiens 27-32 33672092-6 2021 We summarize the association findings between drug hypersensitivity reactions and variants in the genes that encode the enzymes related to the redox system such as enzymes related to glutathione: Glutathione S-transferase (GSTM1, GSTP, GSTT1) and glutathione peroxidase (GPX1), thioredoxin reductase (TXNRD1 and TXNRD2), superoxide dismutase (SOD1, SOD2, and SOD3), catalase (CAT), aldo-keto reductase (AKR), and the peroxiredoxin system (PRDX1, PRDX2, PRDX3, PRDX4, PRDX5, PRDX6). Glutathione 183-194 glutathione S-transferase theta 1 Homo sapiens 236-241 33546147-0 2021 Associations of Metabolic Genes (GSTT1, GSTP1, GSTM1) and Blood Mercury Concentrations Differ in Jamaican Children with and without Autism Spectrum Disorder. Mercury 64-71 glutathione S-transferase theta 1 Homo sapiens 33-38 33544514-9 2021 CONCLUSIONS: Our findings suggest that GSTT1 null genotype may be associated with a decreased risk for pleural plaques and asbestosis, may modify the association between asbestos exposure and MM and may consequently act protectively on MM risk. Asbestos 123-131 glutathione S-transferase theta 1 Homo sapiens 39-44 33381973-0 2021 Effects of GSTT1 Genotype on the Detoxification of 1,3-Butadiene Derived Diepoxide and Formation of Promutagenic DNA-DNA Cross-Links in Human Hapmap Cell Lines. 1,3-butadiene 51-64 glutathione S-transferase theta 1 Homo sapiens 11-16 33381973-0 2021 Effects of GSTT1 Genotype on the Detoxification of 1,3-Butadiene Derived Diepoxide and Formation of Promutagenic DNA-DNA Cross-Links in Human Hapmap Cell Lines. diepoxide 73-82 glutathione S-transferase theta 1 Homo sapiens 11-16 33381973-6 2021 Glutathione-S-transferase theta 1 (GSTT1) facilitates the conjugation of DEB to glutathione as the first step of its detoxification and subsequent elimination via the mercapturic acid pathway. Glutathione 80-91 glutathione S-transferase theta 1 Homo sapiens 0-33 33381973-6 2021 Glutathione-S-transferase theta 1 (GSTT1) facilitates the conjugation of DEB to glutathione as the first step of its detoxification and subsequent elimination via the mercapturic acid pathway. Glutathione 80-91 glutathione S-transferase theta 1 Homo sapiens 35-40 33381973-6 2021 Glutathione-S-transferase theta 1 (GSTT1) facilitates the conjugation of DEB to glutathione as the first step of its detoxification and subsequent elimination via the mercapturic acid pathway. Acetylcysteine 167-183 glutathione S-transferase theta 1 Homo sapiens 0-33 33381973-6 2021 Glutathione-S-transferase theta 1 (GSTT1) facilitates the conjugation of DEB to glutathione as the first step of its detoxification and subsequent elimination via the mercapturic acid pathway. Acetylcysteine 167-183 glutathione S-transferase theta 1 Homo sapiens 35-40 33381973-7 2021 Human biomonitoring studies have revealed a strong association between GSTT1 copy number and urinary concentrations of BD-mercapturic acids, suggesting that it plays an important role in the metabolism of BD. bd-mercapturic acids 119-139 glutathione S-transferase theta 1 Homo sapiens 71-76 33381973-11 2021 Consistent with the protective effect of GSH conjugation against DEB-derived apoptosis, GSTT1 positive cell lines formed significantly more DEB-GSH conjugate than GSTT1 negative cell lines. Glutathione 41-44 glutathione S-transferase theta 1 Homo sapiens 88-93 33381973-11 2021 Consistent with the protective effect of GSH conjugation against DEB-derived apoptosis, GSTT1 positive cell lines formed significantly more DEB-GSH conjugate than GSTT1 negative cell lines. Glutathione 41-44 glutathione S-transferase theta 1 Homo sapiens 163-168 33381973-11 2021 Consistent with the protective effect of GSH conjugation against DEB-derived apoptosis, GSTT1 positive cell lines formed significantly more DEB-GSH conjugate than GSTT1 negative cell lines. Glutathione 144-147 glutathione S-transferase theta 1 Homo sapiens 88-93 32719018-10 2020 CONCLUSIONS: The polymorphism in GSTM1 and GSTT1 gene significantly damage DNA in personnel occupationally exposed to VA. Vanillic Acid 118-120 glutathione S-transferase theta 1 Homo sapiens 43-48 32734539-1 2020 The objective of this study was to identify and evaluate the impact of exposure to mixtures of organochloride pesticides (OCPs) in agricultural workers by detecting their effects on the activity of the enzyme glutathione S-transferase (GST) and the presence of polymorphisms of the GSTT1 and GSTM1 genes. organochloride 95-109 glutathione S-transferase theta 1 Homo sapiens 282-287 33452993-0 2021 Effects of GSTT1 and GSTM1 polymorphisms in glutathione levels and breast cancer development in Brazilian patients. Glutathione 44-55 glutathione S-transferase theta 1 Homo sapiens 11-16 33452993-6 2021 The mean concentration values in nmol/L of GSH were 20.37 +- 5.82 for patients with null genotypes for both genes, 19.75 +- 3.47 for null GSTT1, 17.22 +- 1.35 for active GSTT1, 18.82 +- 1.96 for absent GSTM1, and 16.59 +- 1.66 for active GSTM1, but no significance was found. Glutathione 43-46 glutathione S-transferase theta 1 Homo sapiens 170-175 33834724-6 2021 RESULTS: In the insomnia group, carriers of the normal GSTT1 genotype had higher diene conjugates with lower glutathione peroxidase activity as compared to controls. diene 81-86 glutathione S-transferase theta 1 Homo sapiens 55-60 33834724-6 2021 RESULTS: In the insomnia group, carriers of the normal GSTT1 genotype had higher diene conjugates with lower glutathione peroxidase activity as compared to controls. Glutathione 109-120 glutathione S-transferase theta 1 Homo sapiens 55-60 33834724-7 2021 When comparing groups of women with the deletion in GSTT1, an increase in the content of substrates and lipid peroxidation products was observed at all stages of lipid peroxidation, oxidized glutathione, glutathione S-transferase activity with a decrease in the content of reduced glutathione and retinol in the insomnia group as compared to controls. Glutathione 191-202 glutathione S-transferase theta 1 Homo sapiens 52-57 33834724-7 2021 When comparing groups of women with the deletion in GSTT1, an increase in the content of substrates and lipid peroxidation products was observed at all stages of lipid peroxidation, oxidized glutathione, glutathione S-transferase activity with a decrease in the content of reduced glutathione and retinol in the insomnia group as compared to controls. Glutathione 204-215 glutathione S-transferase theta 1 Homo sapiens 52-57 33834724-7 2021 When comparing groups of women with the deletion in GSTT1, an increase in the content of substrates and lipid peroxidation products was observed at all stages of lipid peroxidation, oxidized glutathione, glutathione S-transferase activity with a decrease in the content of reduced glutathione and retinol in the insomnia group as compared to controls. Vitamin A 297-304 glutathione S-transferase theta 1 Homo sapiens 52-57 32289338-8 2020 However, in children with GSTM1 null and GSTT1 present, increasing acetaminophen use for non-respiratory reasons was associated with reduced FEV1 and MEF at 18 years (interaction between GSTM1/T1 and acetaminophen p<0 05). Acetaminophen 67-80 glutathione S-transferase theta 1 Homo sapiens 41-46 32661462-7 2020 We used conditional logistic regression (CLR) models to assess associations of PCBs and OC pesticides with ASD, individually or interactively with GST genes (GSTT1, GSTM1, GSTP1). Polychlorinated Biphenyls 79-83 glutathione S-transferase theta 1 Homo sapiens 158-163 32022258-11 2020 For example, higher dietary iron was most strongly associated with increased breast cancer risk among women with GSTT1 deletion or GSTM1/GSTT1 double deletions (pinteraction < 0.05). Iron 28-32 glutathione S-transferase theta 1 Homo sapiens 113-118 32022258-11 2020 For example, higher dietary iron was most strongly associated with increased breast cancer risk among women with GSTT1 deletion or GSTM1/GSTT1 double deletions (pinteraction < 0.05). Iron 28-32 glutathione S-transferase theta 1 Homo sapiens 137-142 33295410-1 2020 BACKGROUND: This study aimed to investigate the deletion polymorphisms of the genes of the glutathione S-transferase family GSTT1 and GSTM1 in patients with Polycystic Ovarian Syndrome (PCOS), comparing them with a control population. Glutathione 91-102 glutathione S-transferase theta 1 Homo sapiens 124-129 32525579-9 2020 Also, the GSTT1 null genotype was associated with increased TBARS in oligozoospermia and asthenozoospermia. Thiobarbituric Acid Reactive Substances 60-65 glutathione S-transferase theta 1 Homo sapiens 10-15 32488710-3 2020 Genetic polymorphisms in genes encoding PAH-metabolizing enzymes like glutathione S-transferases (GSTM1, GSTP1, GSTT1) which conjugate glutathione to PAHs for reduction of oxidative stress may affect an individual"s response to PAH exposure. Polycyclic Aromatic Hydrocarbons 40-43 glutathione S-transferase theta 1 Homo sapiens 112-117 32488710-3 2020 Genetic polymorphisms in genes encoding PAH-metabolizing enzymes like glutathione S-transferases (GSTM1, GSTP1, GSTT1) which conjugate glutathione to PAHs for reduction of oxidative stress may affect an individual"s response to PAH exposure. Glutathione 70-81 glutathione S-transferase theta 1 Homo sapiens 112-117 32488710-3 2020 Genetic polymorphisms in genes encoding PAH-metabolizing enzymes like glutathione S-transferases (GSTM1, GSTP1, GSTT1) which conjugate glutathione to PAHs for reduction of oxidative stress may affect an individual"s response to PAH exposure. Glutathione 135-146 glutathione S-transferase theta 1 Homo sapiens 112-117 32488710-3 2020 Genetic polymorphisms in genes encoding PAH-metabolizing enzymes like glutathione S-transferases (GSTM1, GSTP1, GSTT1) which conjugate glutathione to PAHs for reduction of oxidative stress may affect an individual"s response to PAH exposure. Polycyclic Aromatic Hydrocarbons 150-153 glutathione S-transferase theta 1 Homo sapiens 112-117 32488710-10 2020 The null state of GSTT1 was significantly associated with high concentrations of 1-OHP in urea (p < 0.01). Oxaliplatin 81-86 glutathione S-transferase theta 1 Homo sapiens 18-23 32488710-10 2020 The null state of GSTT1 was significantly associated with high concentrations of 1-OHP in urea (p < 0.01). Urea 90-94 glutathione S-transferase theta 1 Homo sapiens 18-23 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. 3,4-epoxy-1-butene 33-51 glutathione S-transferase theta 1 Homo sapiens 85-118 32751086-5 2020 Moreover, serum vitamin C level also is dependent on genetic factors, such as SLC23A1 and SLC23A2 genes, encoding sodium-dependent vitamin C transporters and GSTM1, GSTP1 and GSTT1 genes which encode glutathione S-transferases. Ascorbic Acid 16-25 glutathione S-transferase theta 1 Homo sapiens 175-180 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. 3,4-epoxy-1-butene 33-51 glutathione S-transferase theta 1 Homo sapiens 120-125 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. 3,4-epoxy-1-butene 53-55 glutathione S-transferase theta 1 Homo sapiens 85-118 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. 3,4-epoxy-1-butene 53-55 glutathione S-transferase theta 1 Homo sapiens 120-125 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. Glutathione 85-96 glutathione S-transferase theta 1 Homo sapiens 120-125 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. N-7-(1-hydroxy-3-buten-2-yl)guanine 196-230 glutathione S-transferase theta 1 Homo sapiens 85-118 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. N-7-(1-hydroxy-3-buten-2-yl)guanine 196-230 glutathione S-transferase theta 1 Homo sapiens 120-125 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. eb-gii 232-238 glutathione S-transferase theta 1 Homo sapiens 85-118 32237725-6 2020 BD is metabolically activated to 3,4-epoxy-1-butene (EB), which can be detoxified by glutathione S-transferase theta 1 (GSTT1)-mediated conjugation with glutathione, or can react with DNA to form N7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. eb-gii 232-238 glutathione S-transferase theta 1 Homo sapiens 120-125 32237725-8 2020 We found that GSTT1- HapMap cells treated with EB in culture produced lower levels of glutathione conjugates and were more susceptible to apoptosis, but had similar numbers of EB-GII adducts. Glutathione 86-97 glutathione S-transferase theta 1 Homo sapiens 14-19 32237725-9 2020 Our results suggest that GSTT1 can influence an individual"s susceptibility to butadiene-derived epoxides. butadiene-derived epoxides 79-105 glutathione S-transferase theta 1 Homo sapiens 25-30 32188413-8 2020 Analysis by gender, BMI and alcohol showed that association of GSTT1-null with risk of essential hypertension seems to be significant when BMI < 30 Kg/m2, in non-smokers and in alcohol users (all OR >= 1.77; p <= 0.008). Alcohols 28-35 glutathione S-transferase theta 1 Homo sapiens 63-68 32435918-0 2020 GSTM1 null and GSTT1 null: predictors of cisplatin-caused acute ototoxicity measured by DPOAEs. Cisplatin 41-50 glutathione S-transferase theta 1 Homo sapiens 15-20 32543462-11 2020 Only GSTT1 polymorphism recorded a significant change in percent methylation of Alu elements among urban and rural groups. METHYL HYDROGEN (S)-ACETYLPHOSPHONATE 80-83 glutathione S-transferase theta 1 Homo sapiens 5-10 32188413-8 2020 Analysis by gender, BMI and alcohol showed that association of GSTT1-null with risk of essential hypertension seems to be significant when BMI < 30 Kg/m2, in non-smokers and in alcohol users (all OR >= 1.77; p <= 0.008). Alcohols 177-184 glutathione S-transferase theta 1 Homo sapiens 63-68 32188413-10 2020 CONCLUSION: Our results confirm that GSTT1-null genotype is significantly associated with risk of developing essential hypertension in Burkinabe, especially when BMI < 30 Kg/m2, in non-smokers and in alcohol users, and it showed that the double deletion GSTM1-null/GSTT1-null genotypes may influence body lipids repartition. Alcohols 200-207 glutathione S-transferase theta 1 Homo sapiens 37-42 32342808-8 2020 Subsequently, the levels of, glycolic acid, erythronic acid, lactic acid, citric acid, fructose, stearic acid, 2-amino-2-methyl-1,3-propanediol and three unknown metabolites increased in GSTT1 positive patients; and the levels of proline, valine and two unknown metabolites decreased in GSTT1 positive patients. 2-amino-2-methyl-1,3-propandiol 111-143 glutathione S-transferase theta 1 Homo sapiens 187-192 31916636-1 2020 The enzyme glutathione S-transferase theta 1 (GSTT1) is involved in detoxifying chemicals, including reactive oxygen species (ROS). Reactive Oxygen Species 101-124 glutathione S-transferase theta 1 Homo sapiens 11-44 31916636-1 2020 The enzyme glutathione S-transferase theta 1 (GSTT1) is involved in detoxifying chemicals, including reactive oxygen species (ROS). Reactive Oxygen Species 101-124 glutathione S-transferase theta 1 Homo sapiens 46-51 31916636-1 2020 The enzyme glutathione S-transferase theta 1 (GSTT1) is involved in detoxifying chemicals, including reactive oxygen species (ROS). Reactive Oxygen Species 126-129 glutathione S-transferase theta 1 Homo sapiens 11-44 31916636-1 2020 The enzyme glutathione S-transferase theta 1 (GSTT1) is involved in detoxifying chemicals, including reactive oxygen species (ROS). Reactive Oxygen Species 126-129 glutathione S-transferase theta 1 Homo sapiens 46-51 31771940-9 2020 Much stronger effect was observed among subjects with the null genotype of both GSTM1 and GSTT1: in these individuals, PEITC increased urinary levels of MHBMA by 58.68% (p = 0.004) and 89.97% (p = 0.001), respectively, but did not have a significant effect on urinary DHBMA. phenethyl isothiocyanate 119-124 glutathione S-transferase theta 1 Homo sapiens 90-95 31771940-9 2020 Much stronger effect was observed among subjects with the null genotype of both GSTM1 and GSTT1: in these individuals, PEITC increased urinary levels of MHBMA by 58.68% (p = 0.004) and 89.97% (p = 0.001), respectively, but did not have a significant effect on urinary DHBMA. 1-hydroxybutene-2-yl mercapturic acid 153-158 glutathione S-transferase theta 1 Homo sapiens 90-95 31771940-9 2020 Much stronger effect was observed among subjects with the null genotype of both GSTM1 and GSTT1: in these individuals, PEITC increased urinary levels of MHBMA by 58.68% (p = 0.004) and 89.97% (p = 0.001), respectively, but did not have a significant effect on urinary DHBMA. dhbma 268-273 glutathione S-transferase theta 1 Homo sapiens 90-95 31771940-10 2020 These results reveal a potentially protective effect of PEITC treatment with respect to the detoxification of 1,3-butadiene in cigarette smokers, specifically in those null for GSTT1, and provide further evidence in support of stronger chemopreventive effects from consumption of dietary isothiocyanates in these individuals. phenethyl isothiocyanate 56-61 glutathione S-transferase theta 1 Homo sapiens 177-182 31218944-0 2019 Polymorphisms in GSTM1 and GSTT1 influence the response and treatment outcome in lung cancer patients treated with platinum-based chemotherapy. Platinum 115-123 glutathione S-transferase theta 1 Homo sapiens 27-32 31681592-14 2019 The in vitro study revealed significantly higher number of apoptotic cells at 12 nM of bortezomib in GSTT1-present, GSTM1-null/present, -308GG and -238GG/GA+AA genotypes. bortezomib 87-97 glutathione S-transferase theta 1 Homo sapiens 101-106 30803216-6 2019 Moreover, risk was decreased in alcohol users carrying the GSTT1null (OR=0.39; 95% CI=0.16-0.97) genotype. Alcohols 32-39 glutathione S-transferase theta 1 Homo sapiens 59-64 31249357-2 2019 Glutathione S-transferases (GSTs) participate in CDDP excretion from cells, and genes encoding GSTs, GSTM1, GSTT1and GSTP1, are polymorphic in humans. Cisplatin 49-53 glutathione S-transferase theta 1 Homo sapiens 108-113 31249357-7 2019 Patients with GSTT1 genes showed 7.23- and 5.37-fold higher likelihood of presenting vomiting and ototoxicity, lower glomerular filtration rate (GFR), and lower elimination of CDDP in urine relative to patients with deleted genes. Cisplatin 176-180 glutathione S-transferase theta 1 Homo sapiens 14-19 31249357-10 2019 Our findings provide preliminary evidence that inherited abnormalities in CDDP metabolism, related to GSTT1 and GSTP1 Ile105Val polymorphisms, alter outcomes of HNSCC patients treated with CDDP and RT. Cisplatin 74-78 glutathione S-transferase theta 1 Homo sapiens 102-107 28403014-8 2018 This meta-analysis indicates a possible association between the variant genotypes of GSTM1, GSTT1, NAT2 and SULT1A1, occupational exposure to aromatic amines or PAHs, and development of BC. aromatic amines 142-157 glutathione S-transferase theta 1 Homo sapiens 92-97 30650146-14 2019 After grouping PFOA levels into three groups: undetected, below and above the median in those with detected, children in above the median group who had the GSTT1-null, or GSTM1-null genotype exhibited a higher odds ratio for AD (OR [95%CI] = 3.45 [1.26-9.99] and 2.92 [1.12-7.91], respectively) as compared to the undetected group. perfluorooctanoic acid 15-19 glutathione S-transferase theta 1 Homo sapiens 156-161 30650146-15 2019 CONCLUSIONS: Our data demonstrated that in-utero PFOA exposure with GSTT1/M1 null genotype were associated with AD. perfluorooctanoic acid 49-53 glutathione S-transferase theta 1 Homo sapiens 68-73 30341887-6 2018 RESULTS Twenty-three published studies were identified that showed that both the null GSTM1 and the GG genotype of GSTP1 IIe105Val were associated with improved treatment response to cisplatin-based chemotherapy (GSTT1 present/null: OR=1.328; 95% CI, 1.074-1.643) (GSTP1 GG + AG vs. AA: OR=0.596; 95% CI, 0.468-0.759). Cisplatin 183-192 glutathione S-transferase theta 1 Homo sapiens 213-218 30471640-1 2019 Genetic variations in the glutathione S-transferase genes GSTT1 and GSTM1 have been widely studied, and homozygous deletions or null genotypes have been reported in different populations. Glutathione 26-37 glutathione S-transferase theta 1 Homo sapiens 58-63 28403014-8 2018 This meta-analysis indicates a possible association between the variant genotypes of GSTM1, GSTT1, NAT2 and SULT1A1, occupational exposure to aromatic amines or PAHs, and development of BC. Polycyclic Aromatic Hydrocarbons 161-165 glutathione S-transferase theta 1 Homo sapiens 92-97 29145054-10 2018 For combined analysis with GSTM1 and GSTT1, GSTM1 null and GSTT1 present group showed a significant inverse association of BPb with birthweight and head circumference in males. bpb 123-126 glutathione S-transferase theta 1 Homo sapiens 37-42 29845565-0 2018 Associations between sperm quality, DNA damage, and CYP1A1, GSTT1 and GSTM1 polymorphisms with 1-hydroxypyrene urinary levels in men occupationally exposed to polycyclic aromatic hydrocarbons. 1-hydroxypyrene 95-110 glutathione S-transferase theta 1 Homo sapiens 60-65 29845565-0 2018 Associations between sperm quality, DNA damage, and CYP1A1, GSTT1 and GSTM1 polymorphisms with 1-hydroxypyrene urinary levels in men occupationally exposed to polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 159-191 glutathione S-transferase theta 1 Homo sapiens 60-65 30112115-13 2018 To conclude, deletion of GSTT1, rs1695 and rs1799793 may constitute potential predictors of platinum-induced ototoxicity. Platinum 92-100 glutathione S-transferase theta 1 Homo sapiens 25-30 29875071-9 2018 Recent molecular epidemiology has indicated that activation of brominated trihalomethanes by the enzyme GSTT1 and the lack of metabolism of haloacetic acids by a variant of enzyme GSTZ1 are likely causative mechanisms for bladder cancer associated with exposure to chlorinated water. Trihalomethanes 74-89 glutathione S-transferase theta 1 Homo sapiens 104-109 29204680-0 2018 Association between polymorphism of GSTP1, GSTT1, GSTM1 and CYP2E1 genes and susceptibility to benzene-induced hematotoxicity. Benzene 95-102 glutathione S-transferase theta 1 Homo sapiens 43-48 29204680-3 2018 The main objective of this study was to ascertain whether polymorphism of GSTP1, GSTM1, GSTT1 and CYP2E1 genes might influence susceptibility to the adverse effects of benzene among employees of a petrochemical plant. Benzene 168-175 glutathione S-transferase theta 1 Homo sapiens 88-93 29204680-12 2018 The results of this study showed that, individuals carrying null GSTT1 or both null STT1 and GSTM1 genotypes had a higher risk and were more susceptible to benzene-induced hematological disorders. Benzene 156-163 glutathione S-transferase theta 1 Homo sapiens 65-70 29145054-10 2018 For combined analysis with GSTM1 and GSTT1, GSTM1 null and GSTT1 present group showed a significant inverse association of BPb with birthweight and head circumference in males. bpb 123-126 glutathione S-transferase theta 1 Homo sapiens 59-64 29582627-1 2018 Alcohol detoxification is governed by ADH1B,ALDH2, GSTM1 and GSTT1 genes that encode functional enzymes which are coordinated with each other to removehighly toxic metabolites i.e. acetaldehyde as well as reactive oxygen species generated through detoxification processes.Some communities in the population appears to be at greater risk for development of the liver cancer due to geneticpredispositions. Alcohols 0-7 glutathione S-transferase theta 1 Homo sapiens 61-66 29582627-1 2018 Alcohol detoxification is governed by ADH1B,ALDH2, GSTM1 and GSTT1 genes that encode functional enzymes which are coordinated with each other to removehighly toxic metabolites i.e. acetaldehyde as well as reactive oxygen species generated through detoxification processes.Some communities in the population appears to be at greater risk for development of the liver cancer due to geneticpredispositions. Acetaldehyde 181-193 glutathione S-transferase theta 1 Homo sapiens 61-66 29582627-1 2018 Alcohol detoxification is governed by ADH1B,ALDH2, GSTM1 and GSTT1 genes that encode functional enzymes which are coordinated with each other to removehighly toxic metabolites i.e. acetaldehyde as well as reactive oxygen species generated through detoxification processes.Some communities in the population appears to be at greater risk for development of the liver cancer due to geneticpredispositions. Reactive Oxygen Species 205-228 glutathione S-transferase theta 1 Homo sapiens 61-66 29523098-1 2018 BACKGROUND: The aim of this study was to evaluate whether the glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) null alleles may contribute to carbamazepine-induced hepatotoxicity. Carbamazepine 145-158 glutathione S-transferase theta 1 Homo sapiens 107-112 29523098-2 2018 METHODS: A cross-sectional prospective study was conducted to identify the frequency distribution of GSTM1 and GSTT1 alleles in 129 Tunisian epileptic patients treated with carbamazepine. Carbamazepine 173-186 glutathione S-transferase theta 1 Homo sapiens 111-116 30701758-4 2018 The analysis of the frequency distribution of deletion polymorphisms of GSTM1 and GSTT1 glutathione among the workers of the basic trades involving patients with cardiovascular disease (CVD) and practically healthy workers. Glutathione 88-99 glutathione S-transferase theta 1 Homo sapiens 82-87 30454686-4 2018 In contrast, glutathione-S-transferase (GST) M1 and GSTT1 are primary responsible for detoxification of the AFB1 by catalyzing the conjugation of GSH to AFBO in humans, whereas GSTM2 in a nonhuman primate, GSTA3 in mice, GSTA5 in rats, and GSTA1, GSTA2, GSTA3 and GSTA4 in the turkey are important. Glutathione 146-149 glutathione S-transferase theta 1 Homo sapiens 52-57 28898950-8 2018 When maternal urinary BPA concentration in the third trimester increased by 1 log-transformed unit of BPA/Cr, the third trimester femur length decreased 0.03(0.01)cm in the whole and 0.06(0.02)cm in the GSTM1/GSTT1 either null group. bisphenol A 22-25 glutathione S-transferase theta 1 Homo sapiens 209-214 29619129-6 2018 Independent associations were found between the variant A allele and lower total cholesterol, between c2 allele and low alpha-1 antitrypsin and between the null GSTT1 variant and high indirect and total bilirubin in SCA-HU+ patients. Bilirubin 203-212 glutathione S-transferase theta 1 Homo sapiens 161-166 27168101-4 2017 In patients developing neutropenia, ABCB1 3435TT and homozygosity for GSTT1null (glutathione-S-transferase; conjugates reactive clozapine metabolites into glutathione) were more frequent compared with control (34% versus 20%, P=0.05 and 31% versus 14%, P=0.03), whereas GSTM1null was less frequent in these patients (31% versus 52%, P=0.03). Clozapine 128-137 glutathione S-transferase theta 1 Homo sapiens 70-75 29074540-6 2017 Higher NO2 exposure was associated with lower forced vital capacity for carriers of the GSTT1 null genotype.TRAP exposures were associated with increased risk of asthma, wheeze and lower lung function in middle-aged adults. Nitrogen Dioxide 7-10 glutathione S-transferase theta 1 Homo sapiens 88-93 28386678-7 2017 GSTM1 and LEPR variants exhibited risk modulation for non-muscle-invasive bladder cancer (NMIBC); GSTT1 and PPARG variants for muscle-invasive bladder cancer (MIBC), and ACE variant for NMIBC as well as MIBC. 4-METHYL-2-PENTANOL 159-163 glutathione S-transferase theta 1 Homo sapiens 98-103 28976264-1 2017 AIM: To investigate the impact of genetic variants in CYP2C9, CYP2C19, CYP3A4, GSTT1, GSTM1 and GSTP1 on the efficacy of cyclophosphamide (CYC) therapy in patients with lupus nephritis. Cyclophosphamide 121-137 glutathione S-transferase theta 1 Homo sapiens 79-84 28976264-1 2017 AIM: To investigate the impact of genetic variants in CYP2C9, CYP2C19, CYP3A4, GSTT1, GSTM1 and GSTP1 on the efficacy of cyclophosphamide (CYC) therapy in patients with lupus nephritis. Cyclophosphamide 139-142 glutathione S-transferase theta 1 Homo sapiens 79-84 27168101-4 2017 In patients developing neutropenia, ABCB1 3435TT and homozygosity for GSTT1null (glutathione-S-transferase; conjugates reactive clozapine metabolites into glutathione) were more frequent compared with control (34% versus 20%, P=0.05 and 31% versus 14%, P=0.03), whereas GSTM1null was less frequent in these patients (31% versus 52%, P=0.03). Glutathione 81-92 glutathione S-transferase theta 1 Homo sapiens 70-75 28572675-6 2017 Our meta-analysis suggested that the GSTP1 IIe105Val, GSTM1 and GSTT1 null variants might be predictive factors for the efficacy of platinum-based chemotherapy to NSCLC patients. Platinum 132-140 glutathione S-transferase theta 1 Homo sapiens 64-69 28692121-13 2017 According to the analysis of the variable alcohol consumption, we found that in the case group the presence of the GSTT1 gene was higher in individuals who reported not drinking alcohol. Alcohols 42-49 glutathione S-transferase theta 1 Homo sapiens 115-120 28692121-13 2017 According to the analysis of the variable alcohol consumption, we found that in the case group the presence of the GSTT1 gene was higher in individuals who reported not drinking alcohol. Alcohols 178-185 glutathione S-transferase theta 1 Homo sapiens 115-120 28744640-3 2017 A decrease in glutathione S-transferase activity was found in blood and ejaculate specimens from fertile and infertile carriers of nonfunctional GSTT1(0/0)/GSTM1(0/0) genotypes. Glutathione 14-25 glutathione S-transferase theta 1 Homo sapiens 145-150 28744640-4 2017 In infertile carriers of nonfunctional GSTT1(0/0)/GSTM1(0/0) genotypes determining reduced glutathione S-transferase activity, a decrease in the concentration of low-molecular-weight cell antioxidant (reduced glutathione) and an increase in the concentration of secondary LPO products (TBA-reactive substances) were revealed. Glutathione 91-102 glutathione S-transferase theta 1 Homo sapiens 39-44 28584578-8 2017 Plasma levels of alpha-tocopherol increased significantly in GSTT1 wild genotype (P < 0.05); however, plasma level of beta-carotene increased significantly in GSTT1 null genotype (P < 0.01). alpha-Tocopherol 17-33 glutathione S-transferase theta 1 Homo sapiens 61-66 28584578-8 2017 Plasma levels of alpha-tocopherol increased significantly in GSTT1 wild genotype (P < 0.05); however, plasma level of beta-carotene increased significantly in GSTT1 null genotype (P < 0.01). beta Carotene 121-134 glutathione S-transferase theta 1 Homo sapiens 162-167 28521016-2 2017 Dihaloalkanes are metabolically activated by GSH S-transferase theta1 (GSTT1) to yield products such as episulfonium ions. dihaloalkanes 0-13 glutathione S-transferase theta 1 Homo sapiens 45-69 28521016-2 2017 Dihaloalkanes are metabolically activated by GSH S-transferase theta1 (GSTT1) to yield products such as episulfonium ions. dihaloalkanes 0-13 glutathione S-transferase theta 1 Homo sapiens 71-76 28521016-3 2017 However, whether the GSTT1-mediated step of these dihaloalkanes is related to occupational cholangiocarcinoma is not known. dihaloalkanes 50-63 glutathione S-transferase theta 1 Homo sapiens 21-26 28445029-5 2017 Surprisingly, we also show that fragment-based NHS-ester ligands can be made to confer selectivity for specific lysine hotspots on specific targets including Dpyd, Aldh2, and Gstt1. phloretic acid 47-56 glutathione S-transferase theta 1 Homo sapiens 175-180 28445029-5 2017 Surprisingly, we also show that fragment-based NHS-ester ligands can be made to confer selectivity for specific lysine hotspots on specific targets including Dpyd, Aldh2, and Gstt1. Lysine 112-118 glutathione S-transferase theta 1 Homo sapiens 175-180 28426525-6 2017 CONCLUSIONS: GSTT1 and GSTM1 may modulate DNA damage levels of p53 gene when exposed to polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 88-120 glutathione S-transferase theta 1 Homo sapiens 13-18 28431992-7 2017 Whole genome array CGH technique can be used to identify potential genes, biofunctions and chromosomal abnormalities associated with RSA which is supported by our findings of a number of novel CNVs/genes (22q11.23/GSTT1, 3p22.2/CTDSPL, 6p21.32/HLA, 8p22/MSR1, and 14q32.33/AKT1) and pathways in patients affected with RSA. rabbit sperm membrane autoantigen 133-136 glutathione S-transferase theta 1 Homo sapiens 214-219 28572675-7 2017 The use of GSTP1 IIe105Val, GSTM1 and GSTT1 null polymorphisms as predictive factors of efficacy of personalized platinum-based chemotherapy to NSCLC patients requires further verification with multi-center, multi-ethnic and large-sample-size pharmacogenetic studies. Platinum 113-121 glutathione S-transferase theta 1 Homo sapiens 38-43 28365671-1 2017 The aim of this cross-sectional study was to see whether genetic polymorphisms of the enzymes CYP1A1, GSTM1, and GSTT1 are associated with higher risk of coronary artery disease (CAD) and whether they affect lipid profile in 252 subjects living near a natural gas plant, who are likely to be exposed to polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 303-335 glutathione S-transferase theta 1 Homo sapiens 113-118 28575886-11 2017 The recipients" GSTM1 polymorphism, alone and in combination with donors" GSTM1 and GSTT1, significantly affected the creatinine clearance on discharge day. Creatinine 118-128 glutathione S-transferase theta 1 Homo sapiens 84-89 28365671-1 2017 The aim of this cross-sectional study was to see whether genetic polymorphisms of the enzymes CYP1A1, GSTM1, and GSTT1 are associated with higher risk of coronary artery disease (CAD) and whether they affect lipid profile in 252 subjects living near a natural gas plant, who are likely to be exposed to polycyclic aromatic hydrocarbons (PAHs). Polycyclic Aromatic Hydrocarbons 337-341 glutathione S-transferase theta 1 Homo sapiens 113-118 28365671-6 2017 MANCOVA analysis, which included lipid parameters, glucose, and BMI with sex, age, hypertension and smoking status as covariates, showed a significant difference between the GSTT1*0 and GSTT1*1 allele carriers (p=0.001). Glucose 51-58 glutathione S-transferase theta 1 Homo sapiens 174-179 28365671-6 2017 MANCOVA analysis, which included lipid parameters, glucose, and BMI with sex, age, hypertension and smoking status as covariates, showed a significant difference between the GSTT1*0 and GSTT1*1 allele carriers (p=0.001). Glucose 51-58 glutathione S-transferase theta 1 Homo sapiens 186-191 28365671-7 2017 UNIANCOVA with same covariates showed that total cholesterol and triglyceride levels were significantly higher in GSTT1*1 allele carriers than in GSTT1*0 carriers (p<0.001 and p=0.006, respectively). Cholesterol 49-60 glutathione S-transferase theta 1 Homo sapiens 114-119 28365671-7 2017 UNIANCOVA with same covariates showed that total cholesterol and triglyceride levels were significantly higher in GSTT1*1 allele carriers than in GSTT1*0 carriers (p<0.001 and p=0.006, respectively). Cholesterol 49-60 glutathione S-transferase theta 1 Homo sapiens 146-151 28365671-7 2017 UNIANCOVA with same covariates showed that total cholesterol and triglyceride levels were significantly higher in GSTT1*1 allele carriers than in GSTT1*0 carriers (p<0.001 and p=0.006, respectively). Triglycerides 65-77 glutathione S-transferase theta 1 Homo sapiens 114-119 28043265-0 2016 [Effects of genetic polymorphisms of GSTT1 and GSTM1 on metabolism of Polycyclic aromatic hydrocarbon in coal tar pitch workers]. Polycyclic Aromatic Hydrocarbons 70-101 glutathione S-transferase theta 1 Homo sapiens 37-42 28182092-8 2017 In conclusion, the GSTM1 and GSTT1 null genotypes increased the risk for premalignant and endoscopic gastric lesions, modulated by H. pylori, alcohol, or gastrotoxic drug consumption, while the presence of the GSTP1Val allele seemed to reduce the risk for premalignant lesions. Alcohols 142-149 glutathione S-transferase theta 1 Homo sapiens 29-34 28555163-6 2017 Although GSTT1-mediated bioactivation of dihaloalkanes could be a plausible explanation for the production of reactive metabolites related to carcinogenesis based on previous studies, this catalytic pathway might not mainly contribute to 1,2-DCP-related occupational cholangiocarcinoma. dihaloalkanes 41-54 glutathione S-transferase theta 1 Homo sapiens 9-14 26667829-9 2017 In case-only analysis, GSTT1-null genotype among alcohol users showed elevated risk of hepatotoxicity in HIV-infected individuals (P=0.12, OR=1.36, 95% confidence interval (CI): 0.94-1.97) as compared with GSTT1 genotypes. Alcohols 49-56 glutathione S-transferase theta 1 Homo sapiens 23-28 26667829-9 2017 In case-only analysis, GSTT1-null genotype among alcohol users showed elevated risk of hepatotoxicity in HIV-infected individuals (P=0.12, OR=1.36, 95% confidence interval (CI): 0.94-1.97) as compared with GSTT1 genotypes. Alcohols 49-56 glutathione S-transferase theta 1 Homo sapiens 206-211 28043265-1 2016 Objective: To investigate the influence of genetic polymorphisms of GSTT1 and GSTM1 on metabolism of polycyclic aromatic hydrocarbon (PAHs) in coal tar pitch workers and to explore the effective bio-marker of occupational exposure to coal tar pitch. Polycyclic Aromatic Hydrocarbons 101-132 glutathione S-transferase theta 1 Homo sapiens 68-73 28043265-1 2016 Objective: To investigate the influence of genetic polymorphisms of GSTT1 and GSTM1 on metabolism of polycyclic aromatic hydrocarbon (PAHs) in coal tar pitch workers and to explore the effective bio-marker of occupational exposure to coal tar pitch. Polycyclic Aromatic Hydrocarbons 134-138 glutathione S-transferase theta 1 Homo sapiens 68-73 28043265-9 2016 Conclusion: Carrying positive GSTT1 and GSTM1 promote polycyclic aromatic hydrocarbons in the body"s metabolism. Polycyclic Aromatic Hydrocarbons 54-86 glutathione S-transferase theta 1 Homo sapiens 30-35 27713515-9 2016 However, there were nominally statistically significant interactions for SNPs in acrylamide-metabolizing enzymes: CYP2E1 (rs915906 and rs2480258) and the deletions of GSTM1 and GSTT1. Acrylamide 81-91 glutathione S-transferase theta 1 Homo sapiens 177-182 27275760-9 2016 These GWAS results suggested a possible protective effect of lower GSTT1 copy number variants on the diol epoxide pathway, which was an unexpected result. diol epoxide 101-113 glutathione S-transferase theta 1 Homo sapiens 67-72 27151508-8 2016 Moreover, plasma protein carbonyl level 4h after co-exposure was higher in the individuals who have the GSTT1 null genotype. 4h 40-42 glutathione S-transferase theta 1 Homo sapiens 104-109 27448814-11 2016 Our findings also suggested that GSTT1 and hOGG1 gene polymorphisms might play an important role in the individual risk of As-induced carotid atherosclerosis. Arsenic 123-125 glutathione S-transferase theta 1 Homo sapiens 33-38 27084675-9 2016 The genotype distribution showed significant associations between GSTT1 and the As concentration (log10 iAs, P = 0.01) and metabolite patterns (log10 DMA, P = 0.05) in the urine. Cacodylic Acid 150-153 glutathione S-transferase theta 1 Homo sapiens 66-71 27698905-1 2016 GSTT1 gene plays an important role in detoxification and clearance of reactive oxygen species(ROS). Reactive Oxygen Species 70-93 glutathione S-transferase theta 1 Homo sapiens 0-5 27698905-1 2016 GSTT1 gene plays an important role in detoxification and clearance of reactive oxygen species(ROS). Reactive Oxygen Species 94-97 glutathione S-transferase theta 1 Homo sapiens 0-5 26970590-6 2016 Subjects with GSTT1-present and GSTM1-null genotypes were susceptible to male infertility when exposed to 4-n-OP (OR=14.05, 95% CI=4.78-60.20, P=2.34x10(-5)). 4-octylphenol 106-112 glutathione S-transferase theta 1 Homo sapiens 14-19 26970898-0 2016 Interactions between CYP2E1, GSTZ1 and GSTT1 polymorphisms and exposure to drinking water trihalomethanes and their association with semen quality. drinking water trihalomethanes 75-105 glutathione S-transferase theta 1 Homo sapiens 39-44 26970898-1 2016 Trihalomethanes (THMs) have been reported to be associated with altered semen quality, and this association may be modified by inherited differences in cytochrome P450 (CYP2E1) and glutathione S-transferase (GSTZ1 and GSTT1), which metabolize THMs. Trihalomethanes 0-15 glutathione S-transferase theta 1 Homo sapiens 218-223 26970898-1 2016 Trihalomethanes (THMs) have been reported to be associated with altered semen quality, and this association may be modified by inherited differences in cytochrome P450 (CYP2E1) and glutathione S-transferase (GSTZ1 and GSTT1), which metabolize THMs. Trihalomethanes 17-21 glutathione S-transferase theta 1 Homo sapiens 218-223 26970898-5 2016 GSTT1 genotype significantly modified the association between exposure to Br-THMs (sum of BDCM, DBCM and TBM) and below-reference sperm motility (Pint=0.02). br-thms 74-81 glutathione S-transferase theta 1 Homo sapiens 0-5 26970898-6 2016 Men with above-median blood Br-THM levels had an increased odds ratio (OR) of below-reference sperm compared to men with below-median blood Br-THM levels (OR=2.15, 95% CI: 1.11, 4.19) in the GSTT1 null genotype only. br-thm 28-34 glutathione S-transferase theta 1 Homo sapiens 191-196 26970898-8 2016 Our results suggest that GSTT1 polymorphisms modify Br-THM exposure relation with semen quality, and CYP2E1 polymorphisms are associated with internal levels of exposure to THMs. br-thm 52-58 glutathione S-transferase theta 1 Homo sapiens 25-30 26729828-3 2016 Genetic evaluation determined that he had the GSTT1-null and GSTM1-null genotype, known to be an independent risk factor for developing oxaliplatin-induced SOS. Oxaliplatin 136-147 glutathione S-transferase theta 1 Homo sapiens 46-51 27141907-6 2016 Cytochrome P450 (CYP450s) and glutathione S-transferases (GSTs) are the main metabolic enzymes of PAH and many other xenobiotics, and the polymorphisms of CYP1A1, CYP2E1, GSTM1, GSTT1 and GSTT2 may be associated with the risk of preterm birth. Polycyclic Aromatic Hydrocarbons 98-101 glutathione S-transferase theta 1 Homo sapiens 178-183 26959369-0 2016 Benzene Uptake and Glutathione S-transferase T1 Status as Determinants of S-Phenylmercapturic Acid in Cigarette Smokers in the Multiethnic Cohort. S-phenyl-N-acetylcysteine 74-98 glutathione S-transferase theta 1 Homo sapiens 19-47 25990411-11 2016 Individuals with GSTM1-/GSTT1- have a higher susceptibility to NAION (P < 0.001); the GSTM1-/GSTT1+ genotype also had a significantly higher frequency in patients than in controls (P = 0.004). naion 63-68 glutathione S-transferase theta 1 Homo sapiens 24-29 25990411-11 2016 Individuals with GSTM1-/GSTT1- have a higher susceptibility to NAION (P < 0.001); the GSTM1-/GSTT1+ genotype also had a significantly higher frequency in patients than in controls (P = 0.004). naion 63-68 glutathione S-transferase theta 1 Homo sapiens 96-101 27844021-5 2016 The aim of the present study was to investigate the association between GSTM1 and GSTT1 polymorphisms and risk of dependency to opium sap. opium sap 128-137 glutathione S-transferase theta 1 Homo sapiens 82-87 26554337-4 2015 Therefore, we incubated GSTA1, GSTT1, GSTM1, and GSTP1 with glutathione and BO and quantified the formation of S-phenylglutathione. Glutathione 60-71 glutathione S-transferase theta 1 Homo sapiens 31-36 26774148-9 2016 Our study has demonstrated an association of early onset of CDDP induced ototoxicity with the presence of two copies of GSTT1 gene (p=0,009) and with T allele of rs9332377 polymorphism in COMT gene (p=0,001). Cisplatin 60-64 glutathione S-transferase theta 1 Homo sapiens 120-125 26554337-4 2015 Therefore, we incubated GSTA1, GSTT1, GSTM1, and GSTP1 with glutathione and BO and quantified the formation of S-phenylglutathione. benzene oxide 76-78 glutathione S-transferase theta 1 Homo sapiens 31-36 26554337-8 2015 We conclude that GSTT1 is a critical enzyme in the detoxification of BO and that GSTP1 may also play an important role, while GSTA1 and GSTM1 seem to be less important. benzene oxide 69-71 glutathione S-transferase theta 1 Homo sapiens 17-22 26318115-9 2015 The adverse genotype combination (unusual BCHE variants, PON1 55MM/-108TT and null genotype for both GSTM1 and GSTT1) potentially conferring a greater genetic risk from exposure to organophosphates was observed in 0.2% of our study population. Organophosphates 181-197 glutathione S-transferase theta 1 Homo sapiens 111-116 26498776-11 2015 Additionally, Gsta3, Gstm2 and Gstt1 in Burn-CLP were significantly enriched in glutathione metabolism. Glutathione 80-91 glutathione S-transferase theta 1 Homo sapiens 31-36 26877838-1 2015 The aim of this study was to investigate whether genetic polymorphisms of CYP2E1, GSTM1, and GSTT1 and lifestyle habits (smoking, drinking, and exercise) modulate the levels of urinary styrene metabolites such as mandelic acid (MA) and phenylglyoxylic acid (PGA) after occupational exposure to styrene. Styrene 185-192 glutathione S-transferase theta 1 Homo sapiens 93-98 26877838-1 2015 The aim of this study was to investigate whether genetic polymorphisms of CYP2E1, GSTM1, and GSTT1 and lifestyle habits (smoking, drinking, and exercise) modulate the levels of urinary styrene metabolites such as mandelic acid (MA) and phenylglyoxylic acid (PGA) after occupational exposure to styrene. mandelic acid 213-226 glutathione S-transferase theta 1 Homo sapiens 93-98 26877838-1 2015 The aim of this study was to investigate whether genetic polymorphisms of CYP2E1, GSTM1, and GSTT1 and lifestyle habits (smoking, drinking, and exercise) modulate the levels of urinary styrene metabolites such as mandelic acid (MA) and phenylglyoxylic acid (PGA) after occupational exposure to styrene. phenylglyoxylic acid 236-256 glutathione S-transferase theta 1 Homo sapiens 93-98 26877838-1 2015 The aim of this study was to investigate whether genetic polymorphisms of CYP2E1, GSTM1, and GSTT1 and lifestyle habits (smoking, drinking, and exercise) modulate the levels of urinary styrene metabolites such as mandelic acid (MA) and phenylglyoxylic acid (PGA) after occupational exposure to styrene. phenylglyoxylic acid 258-261 glutathione S-transferase theta 1 Homo sapiens 93-98 26877838-1 2015 The aim of this study was to investigate whether genetic polymorphisms of CYP2E1, GSTM1, and GSTT1 and lifestyle habits (smoking, drinking, and exercise) modulate the levels of urinary styrene metabolites such as mandelic acid (MA) and phenylglyoxylic acid (PGA) after occupational exposure to styrene. Styrene 294-301 glutathione S-transferase theta 1 Homo sapiens 93-98 25876999-0 2015 Associations between the polymorphisms of GSTT1, GSTM1 and methylation of arsenic in the residents exposed to low-level arsenic in drinking water in China. Arsenic 74-81 glutathione S-transferase theta 1 Homo sapiens 42-47 26175060-2 2015 In the present study, we confirmed associations between schizophrenia and the common CNVs in the glutathione (GSH)-related genes GSTT1, DDTL, and GSTM1 using quantitative real-time polymerase chain reaction analyses of 620 patients with schizophrenia and in 622 controls. Glutathione 97-108 glutathione S-transferase theta 1 Homo sapiens 129-134 26175060-2 2015 In the present study, we confirmed associations between schizophrenia and the common CNVs in the glutathione (GSH)-related genes GSTT1, DDTL, and GSTM1 using quantitative real-time polymerase chain reaction analyses of 620 patients with schizophrenia and in 622 controls. Glutathione 110-113 glutathione S-transferase theta 1 Homo sapiens 129-134 25595865-5 2015 RESULTS: GSTM1 and GSTT1 deletion was found in 57% (53/93) and 18% (17/93), respectively, in healthy patients, while the OSCC group showed 57% (57/100) for GSTM1 deletion and 22% (22/100) with a deletion of GSTT1. oscc 121-125 glutathione S-transferase theta 1 Homo sapiens 207-212 26404360-10 2015 The subjects with APOE rs429358 T/C + C/C and GSTT1- genotype were found to have the highest plasma TG level, erythrocyte CAT enzyme activity, and the lowest GST enzyme activity compared to subjects with other genotypes (p < 0.05). Triglycerides 100-102 glutathione S-transferase theta 1 Homo sapiens 46-51 26003511-0 2015 Association between null alleles of GSTM1 and GSTT1 and dependence to heroin and opium. Heroin 70-76 glutathione S-transferase theta 1 Homo sapiens 46-51 26295386-0 2015 Genetic Polymorphisms of Glutathione-Related Enzymes (GSTM1, GSTT1, and GSTP1) and Schizophrenia Risk: A Meta-Analysis. Glutathione 25-36 glutathione S-transferase theta 1 Homo sapiens 61-66 26179485-7 2015 White blood cell counts were also lower in workers with null-GSTT1 and null-GSTM after adjusting for age, gender, smoking, and alcohol consumption. Alcohols 127-134 glutathione S-transferase theta 1 Homo sapiens 61-66 26179485-8 2015 CONCLUSION: Null-GSTT1 and null-GSTM1 genotypes and Cytochrome P4502E1 (CYP2E1: rs2031920, rs3813867) may support the hematotoxicity of benzene-exposed workers in China, and we can make use of it to select susceptible population. Benzene 136-143 glutathione S-transferase theta 1 Homo sapiens 17-22 25876999-0 2015 Associations between the polymorphisms of GSTT1, GSTM1 and methylation of arsenic in the residents exposed to low-level arsenic in drinking water in China. Arsenic 120-127 glutathione S-transferase theta 1 Homo sapiens 42-47 25876999-0 2015 Associations between the polymorphisms of GSTT1, GSTM1 and methylation of arsenic in the residents exposed to low-level arsenic in drinking water in China. Water 140-145 glutathione S-transferase theta 1 Homo sapiens 42-47 25876999-1 2015 We carry out a study to analyze the relation between polymorphisms of GSTT1, GSTM1 and the capacity of arsenic methylation in a human population exposed to arsenic in drinking water. Arsenic 103-110 glutathione S-transferase theta 1 Homo sapiens 70-75 25876999-1 2015 We carry out a study to analyze the relation between polymorphisms of GSTT1, GSTM1 and the capacity of arsenic methylation in a human population exposed to arsenic in drinking water. Arsenic 156-163 glutathione S-transferase theta 1 Homo sapiens 70-75 25876999-2 2015 230 randomly chose subjects were divided into four subgroups based on the arsenic levels, and then the associations between the polymorphisms of GSTT1, GSTM1 and methylation of arsenic were investigated. Arsenic 177-184 glutathione S-transferase theta 1 Homo sapiens 145-150 25876999-4 2015 Moreover, the levels of iAs and TAs in urine in the subjects with genotype of GSTM1(+) were significantly higher than those with GSTM1(-); the level of DMA in the subjects with GSTT1(+) and GSTM1(+) were higher than those with GSTT1(-) and GSTM1(-), although it is not statistically significant. dma 152-155 glutathione S-transferase theta 1 Homo sapiens 177-182 25876999-6 2015 The levels of TAs in urine, together with the genotypes of GSTT1/GSTM1 were associated with the levels of MMA and DMA. mma 106-109 glutathione S-transferase theta 1 Homo sapiens 59-64 25876999-6 2015 The levels of TAs in urine, together with the genotypes of GSTT1/GSTM1 were associated with the levels of MMA and DMA. dma 114-117 glutathione S-transferase theta 1 Homo sapiens 59-64 25876999-7 2015 Our results suggested that the polymorphisms of GSTT1 and GSTM1 were associated with the methylation of arsenic, especially the levels of DMA and SMI. Arsenic 104-111 glutathione S-transferase theta 1 Homo sapiens 48-53 25876999-7 2015 Our results suggested that the polymorphisms of GSTT1 and GSTM1 were associated with the methylation of arsenic, especially the levels of DMA and SMI. dma 138-141 glutathione S-transferase theta 1 Homo sapiens 48-53 25648260-4 2015 Cross-talk was observed between one-carbon and xenobiotic pathways in breast cancer (RFC 80 G>A, COMT H108L and TYMS 5"-UTR 28 bp tandem repeat) and SLE (CYP1A1 m1, MTRR 66 A>G and GSTT1). Carbon 36-42 glutathione S-transferase theta 1 Homo sapiens 181-186 25562543-2 2015 The aim of this work was to investigate the effects of polymorphic genes CYP2E1, EPHX1, GSTT1, and GSTM1 on the urinary concentrations of the styrene metabolites mandelic acid (MA), phenylglyoxylic acid (PGA) and on the concentration ratios between (MA+PGA) and urinary styrene (U-Sty) and airborne styrene (A-Sty), in 30 workers from two fiberglass-reinforced plastic manufacturing plants and 26 unexposed controls. Styrene 142-149 glutathione S-transferase theta 1 Homo sapiens 88-93 26258094-2 2015 Detoxification of ROS is largely performed by Glutathione S-transferases (GSTs), therefore polymorphisms of GSTM1, GSTT1 and GSTP1 genes which decrease enzymes activity could affect SLE susceptibility. Reactive Oxygen Species 18-21 glutathione S-transferase theta 1 Homo sapiens 115-120 25562543-2 2015 The aim of this work was to investigate the effects of polymorphic genes CYP2E1, EPHX1, GSTT1, and GSTM1 on the urinary concentrations of the styrene metabolites mandelic acid (MA), phenylglyoxylic acid (PGA) and on the concentration ratios between (MA+PGA) and urinary styrene (U-Sty) and airborne styrene (A-Sty), in 30 workers from two fiberglass-reinforced plastic manufacturing plants and 26 unexposed controls. mandelic acid 162-175 glutathione S-transferase theta 1 Homo sapiens 88-93 26604430-6 2015 We report that GSTT1 null genotype is significantly associated with the risk of CD (OR: 2.5, CI: 1.2-5, P = 0.013) and UC (OR: 3.5, CI: 1.5-8.5, P = 0.004) and can influence Crohn"s disease behavior. Cadmium 80-82 glutathione S-transferase theta 1 Homo sapiens 15-20 25678751-5 2015 In case of GSTT1 on the other hand, the decrease in diastolic pressure and lymphocyte DNA damage was observed in both null types and present types, but the erythrocyte catalase activity was decreased in GSTT1-null type and the plasma vitamin C level was increased in GSTT1-present type, suggesting that, the antioxidant effect of grape juice was greater in GSTT1-present type compared to GSTT1-null type. Ascorbic Acid 234-243 glutathione S-transferase theta 1 Homo sapiens 11-16 25874030-4 2015 We analyzed the correlation between toluene metabolism and genetic diversity in glutathione S-transferase (GST) (M1), GSTT1, and cytochrome p-450 (CYP) 2E1*5 as well as lifestyle habits (smoking, drinking, and exercise habits). Toluene 36-43 glutathione S-transferase theta 1 Homo sapiens 118-123 25874030-5 2015 The results revealed significant correlations between toluene metabolism and GSTM1 and GSTT1 genetic diversity, as well as smoking and exercise. Toluene 54-61 glutathione S-transferase theta 1 Homo sapiens 87-92 25725180-4 2015 Genotypes of GSTM1 and GSTT1 null deletions were determined in 22 (53.7%) and 7 (17.1%) patients with PCAG and 34 (34%) and 15 (15%) in healthy participants. pcag 102-106 glutathione S-transferase theta 1 Homo sapiens 23-28 25725180-5 2015 Comparison of patients and healthy ones regarding GSTM1 and GSTT1 genotypes revealed increase of GSTM1 null deletions genotypes" in patients with PCAG (P=0.03). pcag 146-150 glutathione S-transferase theta 1 Homo sapiens 60-65 25432281-0 2014 Role of glutathione-S-transferase M1 (GSTM1) and T1 (GSTT1) genes in the development and progress of chronic myeloid leukemia and in the formation of response to imatinib therapy. Imatinib Mesylate 162-170 glutathione S-transferase theta 1 Homo sapiens 53-58 27843993-0 2015 Association between GSTM1 and GSTT1 polymorphisms and susceptibility to methamphetamine dependence. Methamphetamine 72-87 glutathione S-transferase theta 1 Homo sapiens 30-35 27843993-4 2015 The aim of the present study is to investigate the association between GSTM1 and GSTT1 polymorphisms and methamphetamine dependence. Methamphetamine 105-120 glutathione S-transferase theta 1 Homo sapiens 81-86 25432281-3 2014 Combinations of the "zero" GSTM1 and GSTT1 genotypes were risk factors indicating the probable disease progress and failure of high cytogenetic response after 12 months of imatinib therapy (400 mg daily). Imatinib Mesylate 172-180 glutathione S-transferase theta 1 Homo sapiens 37-42 25188725-0 2014 Dual glutathione-S-transferase-theta1 and -mu1 gene deletions determine imatinib failure in chronic myeloid leukemia. Imatinib Mesylate 72-80 glutathione S-transferase theta 1 Homo sapiens 5-46 25188725-4 2014 Deletion of GSTT1 alone, or in combination with deletion of the GSTM1 gene, significantly increased the likelihood of imatinib failure (P = 0.021 and P < 0.001, respectively). Imatinib Mesylate 118-126 glutathione S-transferase theta 1 Homo sapiens 12-17 25188725-6 2014 Losses of the GSTT1 and GSTM1 genes are therefore important determinants of imatinib failure in CML. Imatinib Mesylate 76-84 glutathione S-transferase theta 1 Homo sapiens 14-19 25447454-5 2014 As to susceptibility biomarkers, functional genetic polymorphisms of relevant biotransformation enzymes may modulate the risk of adverse effects (NQO1) and the levels of biomarkers of internal dose, in particular S-phenylmercapturic acid (GSTM1, GSTT1, GSTA1). S-phenyl-N-acetylcysteine 213-237 glutathione S-transferase theta 1 Homo sapiens 246-251 24968062-5 2014 GSTT1 null polymorphism reduces the conjugation rate of benzene epoxide with GSH, and to a lesser extent also GSTTA1 mutant, GSTM1 null and NQO1 mutant genotypes. benzene epoxide 56-71 glutathione S-transferase theta 1 Homo sapiens 0-5 24968062-5 2014 GSTT1 null polymorphism reduces the conjugation rate of benzene epoxide with GSH, and to a lesser extent also GSTTA1 mutant, GSTM1 null and NQO1 mutant genotypes. Glutathione 77-80 glutathione S-transferase theta 1 Homo sapiens 0-5 25368399-10 2014 CONCLUSIONS: The difference in urinary MHBMA excretion levels from cigarette smoking across three ethnic groups is, in part, explained by the GSTT1 genotype. mhbma 39-44 glutathione S-transferase theta 1 Homo sapiens 142-147 25420021-10 2014 RESULTS: Individual GSTM1 or GSTT1 gene deletion affects body antioxidant biomarkers levels, including erythrocyte GST activity, plasma total antioxidant capacity, and glutathione levels. Glutathione 168-179 glutathione S-transferase theta 1 Homo sapiens 29-34 25368399-11 2014 Mean urinary MHBMA levels are higher in whites among GSTT1-null smokers. mhbma 13-18 glutathione S-transferase theta 1 Homo sapiens 53-58 24913818-7 2014 Excretion of the benzene-derived mercapturic acid was higher in participants who were GSTT1-positive than in the null genotype, irrespective of study arm assignment. Benzene 17-24 glutathione S-transferase theta 1 Homo sapiens 86-91 25165394-2 2014 Glutathione S-transferase (GST) M1 and GSTT1 metabolize isothiocyanates; genetic variants may result in differences in biologic response. Isothiocyanates 56-71 glutathione S-transferase theta 1 Homo sapiens 39-44 24913818-7 2014 Excretion of the benzene-derived mercapturic acid was higher in participants who were GSTT1-positive than in the null genotype, irrespective of study arm assignment. Acetylcysteine 33-49 glutathione S-transferase theta 1 Homo sapiens 86-91 24637631-2 2014 The primary hypothesis of this study was to identify whether the polymorphisms of two glutathione-S-transferase enzymes (GSTM1 and GSTT1 genes) predict an increased risk of mood and anxiety disorders in smokers with nicotine dependence. Nicotine 216-224 glutathione S-transferase theta 1 Homo sapiens 131-136 24913811-4 2014 The present study aimed to examine the impact of GSTT1 and GSTM1 polymorphisms on the response to imatinib in patients with CML. Imatinib Mesylate 98-106 glutathione S-transferase theta 1 Homo sapiens 49-54 24913811-10 2014 Moreover, the GSTT1 present/GSTM1 present appeared to be associated with a final dose of 600 or 800 mg of imatinib, but not significantly. Imatinib Mesylate 106-114 glutathione S-transferase theta 1 Homo sapiens 14-19 30514013-2 2014 N-acetyltransferase 2 (NAT2), cytochrome P450 2E1 (CYP2E1) and glutathione S-transferase (loci GSTM1 and GSTT1) are involved in the metabolism of isoniazid, the most toxic drug for the treatment of tuberculosis (TB). Isoniazid 146-155 glutathione S-transferase theta 1 Homo sapiens 105-110 24216264-11 2014 To find out the dependence of blood OCPs level on genotype, we carried out logistic regression analysis and results revealed that GSTM1(-)/GSTT1(-) genotype associated significantly with a number of OCPs namely gamma-HCH, p,p"-DDT and total pesticides. Hexachlorocyclohexane 211-220 glutathione S-transferase theta 1 Homo sapiens 139-144 24685594-4 2014 RESULTS: In the cross-sectional study, GSTM1-null genotype and GSTT1-null genotype were associated with EAH in subjects with type 2 diabetes (59.0% vs. 50.3%, p=0.007; 28.5% vs. 20.7%, p=0.008; consequently). eah 104-107 glutathione S-transferase theta 1 Homo sapiens 63-68 24685594-5 2014 CONCLUSION: After adjustment for age, body mass index, and hsCRP level, GSTM1-null and GSTT1-null genotypes were found to be independent risk factors for the development of EAH in Slovenian patients with type 2 diabetes. eah 173-176 glutathione S-transferase theta 1 Homo sapiens 87-92 24593045-7 2014 The GSTT1 deletion was associated with a higher frequency of the NMPA to homozygous deletion (p = 0.008), GSTP1 + 313A > G with a minor risk of osteoporosis (p = 0.036), and patient age <= 154 months (p = 0.044) with the AA genotype. nmpa 65-69 glutathione S-transferase theta 1 Homo sapiens 4-9 24586676-0 2014 GSTT1 deletion is related to polycyclic aromatic hydrocarbons-induced DNA damage and lymphoma progression. Polycyclic Aromatic Hydrocarbons 29-61 glutathione S-transferase theta 1 Homo sapiens 0-5 24586676-3 2014 Here we investigated the molecular connection of the genetic polymorphism of GSTT1 to the response of lymphocytes to polycyclic aromatic hydrocarbons (PAH). Polycyclic Aromatic Hydrocarbons 117-149 glutathione S-transferase theta 1 Homo sapiens 77-82 24586676-3 2014 Here we investigated the molecular connection of the genetic polymorphism of GSTT1 to the response of lymphocytes to polycyclic aromatic hydrocarbons (PAH). Polycyclic Aromatic Hydrocarbons 151-154 glutathione S-transferase theta 1 Homo sapiens 77-82 24586676-6 2014 Mimicking environmental exposure using long-term repeat culture with low-dose PAH metabolite Hydroquinone, malignant B- and T-lymphocytes presented increased DNA damage, pCHK1/MYC expression and cell proliferation, which were counteracted by ectopic expression of GSTT1. Polycyclic Aromatic Hydrocarbons 78-81 glutathione S-transferase theta 1 Homo sapiens 264-269 24586676-6 2014 Mimicking environmental exposure using long-term repeat culture with low-dose PAH metabolite Hydroquinone, malignant B- and T-lymphocytes presented increased DNA damage, pCHK1/MYC expression and cell proliferation, which were counteracted by ectopic expression of GSTT1. hydroquinone 93-105 glutathione S-transferase theta 1 Homo sapiens 264-269 24586676-9 2014 Collectively, these findings suggested that GSTT1 deletion is related to genetic predisposition to lymphoma, particularly interacting with environmental pollutants containing PAH. Polycyclic Aromatic Hydrocarbons 175-178 glutathione S-transferase theta 1 Homo sapiens 44-49 24967061-5 2014 RESULTS: GSTM1 and GSTT1 null deletions genotypes were determined in 22 (53.7%) and 7 (17.1%) patients with PCAG and 34 (34%) and 15 (15%) in healthy subjects. pcag 108-112 glutathione S-transferase theta 1 Homo sapiens 19-24 24967061-6 2014 Comparison between patients and healthy subjects regarding GSTM1 and GSTT1 genotypes revealed increase of GSTM1 null deletions genotypes in patients with PCAG (P=0.03). pcag 154-158 glutathione S-transferase theta 1 Homo sapiens 69-74 24216264-11 2014 To find out the dependence of blood OCPs level on genotype, we carried out logistic regression analysis and results revealed that GSTM1(-)/GSTT1(-) genotype associated significantly with a number of OCPs namely gamma-HCH, p,p"-DDT and total pesticides. DDT 222-230 glutathione S-transferase theta 1 Homo sapiens 139-144 24696865-1 2014 This study aims to evaluate the effects of polymorphisms in glutathione (GSH-) related genes (GSTM1, GSTT1, GSTP1, GCLM, and GCLC) in the distribution of Hg in the blood compartments in humans exposed to methylmercury (MeHg). Glutathione 60-71 glutathione S-transferase theta 1 Homo sapiens 101-106 24696865-1 2014 This study aims to evaluate the effects of polymorphisms in glutathione (GSH-) related genes (GSTM1, GSTT1, GSTP1, GCLM, and GCLC) in the distribution of Hg in the blood compartments in humans exposed to methylmercury (MeHg). Glutathione 73-76 glutathione S-transferase theta 1 Homo sapiens 101-106 25790712-5 2014 CONCLUSION: Children with the neutrophilic phenotype of bronchial asthma having deletions in the GSTT1/GSTM1 system are characterized by bronchial hypersensitivity to histamine and dosed physical exercises. Histamine 167-176 glutathione S-transferase theta 1 Homo sapiens 97-102 25306601-6 2014 Variable sensitivity to dioxins was demonstrated by associations of genetic polymorphism (CYP1A1, GSTM1, GSTT1, n = 195) and congenital morphogenetic variants among children (n = 1734). Dioxins 24-31 glutathione S-transferase theta 1 Homo sapiens 105-110 23979980-1 2014 Glutathione S-transferase T1 (GSTT1) catalyzes reactions between glutathione and lipophilic compounds with electrophilic centers, leading to neutralization of toxic compounds, xenobiotics, and products of oxidative stress. Glutathione 65-76 glutathione S-transferase theta 1 Homo sapiens 0-28 23979980-1 2014 Glutathione S-transferase T1 (GSTT1) catalyzes reactions between glutathione and lipophilic compounds with electrophilic centers, leading to neutralization of toxic compounds, xenobiotics, and products of oxidative stress. Glutathione 65-76 glutathione S-transferase theta 1 Homo sapiens 30-35 23979980-8 2014 In addition, we found that cigarette smoking and alcohol drinking may modified the association of GSTT1 null genotypes with the risk of GC. Alcohols 49-56 glutathione S-transferase theta 1 Homo sapiens 98-103 24908960-3 2014 In patients with neutrophilic BA and deletion polymorphism of genes GSTT1 and GSTM1, there was a tendency to decreasing of the bronchial lability index through the decrease of bronchodilation, and bronchial response to histamine occurred to be higher than in children with the absence of polymorphism of the referred genes of the xenobiotics biotransformation system. Histamine 219-228 glutathione S-transferase theta 1 Homo sapiens 68-73 23888321-1 2014 Genetic polymorphisms in glutathione S-transferases M1 (GSTM1) and T1 (GSTT1) genes have been widely reported and considered to have a significant effect on prostate cancer (PCa) risk, but the results are inconsistent. Glutathione 25-36 glutathione S-transferase theta 1 Homo sapiens 71-76 24098457-10 2013 In diabetic patients, GSTT1-null conferred higher levels of triglycerides and VLDL-cholesterol, while GSTM1-null was associated with increased levels of fasting blood glucose, glycated hemoglobin and blood pressure. Triglycerides 60-73 glutathione S-transferase theta 1 Homo sapiens 22-27 24319713-0 2013 Role of cytochrome P450-dependent monooxygenases and polymorphic variants of GSTT1 and GSTM1 genes in the formation of brain lesions in individuals chronically exposed to mercury. Mercury 171-178 glutathione S-transferase theta 1 Homo sapiens 77-82 24319713-4 2013 Inhibition of antipyrine metabolism, increased frequency of combination of GSTT1(0/0)/GSTM1(+) genotypes in patients with chronic mercury intoxication, and the specificity of cytochrome P450 inhibition with mercury suggest that disease progression is related to inhibition of cytochrome P450 isoforms in the brain that catalyze regulation of endogenous substrates. Mercury 130-137 glutathione S-transferase theta 1 Homo sapiens 75-80 24370292-0 2013 [Association between GSTP1, GSTM1, GSTT1 genetic polymorphisms and urinary styrene phenyl hydroxyethyl mercapturic acids level]. styrene phenyl hydroxyethyl mercapturic acids 75-120 glutathione S-transferase theta 1 Homo sapiens 35-40 24370292-1 2013 OBJECTIVE: To investigate the relationship between genetic polymorphisms of glutathione S-transferase P1 (GSTP1), glutathione S-transferase M1 (GSTM1), and glutathione S-transferase T1 (GSTT1) and urinary level of mercapturic acids of styrene (PHEMAs) in workers exposed to styrene. Acetylcysteine 214-231 glutathione S-transferase theta 1 Homo sapiens 156-184 24370292-1 2013 OBJECTIVE: To investigate the relationship between genetic polymorphisms of glutathione S-transferase P1 (GSTP1), glutathione S-transferase M1 (GSTM1), and glutathione S-transferase T1 (GSTT1) and urinary level of mercapturic acids of styrene (PHEMAs) in workers exposed to styrene. Styrene 235-242 glutathione S-transferase theta 1 Homo sapiens 156-184 24370292-1 2013 OBJECTIVE: To investigate the relationship between genetic polymorphisms of glutathione S-transferase P1 (GSTP1), glutathione S-transferase M1 (GSTM1), and glutathione S-transferase T1 (GSTT1) and urinary level of mercapturic acids of styrene (PHEMAs) in workers exposed to styrene. Polyhydroxyethyl Methacrylate 244-250 glutathione S-transferase theta 1 Homo sapiens 156-184 24370292-1 2013 OBJECTIVE: To investigate the relationship between genetic polymorphisms of glutathione S-transferase P1 (GSTP1), glutathione S-transferase M1 (GSTM1), and glutathione S-transferase T1 (GSTT1) and urinary level of mercapturic acids of styrene (PHEMAs) in workers exposed to styrene. Styrene 274-281 glutathione S-transferase theta 1 Homo sapiens 156-184 24098457-10 2013 In diabetic patients, GSTT1-null conferred higher levels of triglycerides and VLDL-cholesterol, while GSTM1-null was associated with increased levels of fasting blood glucose, glycated hemoglobin and blood pressure. Cholesterol 83-94 glutathione S-transferase theta 1 Homo sapiens 22-27 23827356-4 2013 The aims of the present study was to evaluate the effects of polymorphisms in glutathione (GSH)-related genes (GSTM1, GSTT1, GSTP1 and GCLM) on Hg concentrations in blood and hair, as well as MeHg-related effects on catalase (CAT) and glutathione-peroxidase (GPx) activity and GSH concentrations. Glutathione 91-94 glutathione S-transferase theta 1 Homo sapiens 118-123 24124608-0 2013 GSTT1 copy number gain and ZNF overexpression are predictors of poor response to imatinib in gastrointestinal stromal tumors. Imatinib Mesylate 81-89 glutathione S-transferase theta 1 Homo sapiens 0-5 24124608-7 2013 Surprisingly, all four malignant GISTs with KIT exon 11 deletion mutations with primary resistance to imatinib had an increased GSTT1 CN and mRNA expression level of GSTT1. Imatinib Mesylate 102-110 glutathione S-transferase theta 1 Homo sapiens 128-133 24124608-7 2013 Surprisingly, all four malignant GISTs with KIT exon 11 deletion mutations with primary resistance to imatinib had an increased GSTT1 CN and mRNA expression level of GSTT1. Imatinib Mesylate 102-110 glutathione S-transferase theta 1 Homo sapiens 166-171 24124608-8 2013 Increased mRNA expression of GSTT1 and ZNF could be predictors of a poor response to imatinib. Imatinib Mesylate 85-93 glutathione S-transferase theta 1 Homo sapiens 29-34 23798465-9 2013 The association between AA and GSTT1 deletion suggests a role of glutathione-conjugation in AA, possibly through protecting the hematopoietic compartment from endogenous metabolites or environmental exposures. Glutathione 65-76 glutathione S-transferase theta 1 Homo sapiens 31-36 24511153-7 2013 This data suggests that GSTT1 contributes to the observed variability in arsenic metabolism. Arsenic 73-80 glutathione S-transferase theta 1 Homo sapiens 24-29 23817691-1 2013 Glutathione S-transferases (GSTs) enzymes are involved in conjugation of electrophilic compounds to glutathione, and glutathione S-transferase T 1 (GSTT1) and glutathione S-transferase M 1 (GSTM1) polymorphisms have been implicated as risk factors for prostate cancer. Glutathione 100-111 glutathione S-transferase theta 1 Homo sapiens 148-153 24084344-0 2013 Cytogenetic damage in Turkish coke oven workers exposed to polycyclic aromatic hydrocarbons: Association with CYP1A1, CYP1B1, EPHX1, GSTM1, GSTT1, and GSTP1 gene polymorphisms. Polycyclic Aromatic Hydrocarbons 59-91 glutathione S-transferase theta 1 Homo sapiens 140-145 23810248-12 2013 CONCLUSIONS: The GSTM1-null genotype plays an important role in genetic susceptibility to MIBC and the GSTT1-null genotype is associated with disease progression and shorter survival in MIBC. 4-METHYL-2-PENTANOL 186-190 glutathione S-transferase theta 1 Homo sapiens 103-108 24511153-0 2013 Influence of GSTT1 Genetic Polymorphisms on Arsenic Metabolism. Arsenic 44-51 glutathione S-transferase theta 1 Homo sapiens 13-18 24511153-5 2013 A significant gene-environment interaction was observed between urinary arsenic exposure in drinking water GSTT1 but not GSTM1 where GSTT1 null individuals had a slightly higher excretion rate of arsenic compared to GSTT1 wildtypes after adjusting for other factors. Arsenic 72-79 glutathione S-transferase theta 1 Homo sapiens 107-112 24511153-8 2013 Since individuals with a higher primary methylation index and lower secondary methylation index are more susceptible to arsenic related disease, these results suggest that GSTT1 null individuals may be more susceptible to arsenic-related toxicity. Arsenic 120-127 glutathione S-transferase theta 1 Homo sapiens 172-177 24511153-5 2013 A significant gene-environment interaction was observed between urinary arsenic exposure in drinking water GSTT1 but not GSTM1 where GSTT1 null individuals had a slightly higher excretion rate of arsenic compared to GSTT1 wildtypes after adjusting for other factors. Arsenic 196-203 glutathione S-transferase theta 1 Homo sapiens 133-138 24511153-5 2013 A significant gene-environment interaction was observed between urinary arsenic exposure in drinking water GSTT1 but not GSTM1 where GSTT1 null individuals had a slightly higher excretion rate of arsenic compared to GSTT1 wildtypes after adjusting for other factors. Arsenic 196-203 glutathione S-transferase theta 1 Homo sapiens 133-138 24511153-8 2013 Since individuals with a higher primary methylation index and lower secondary methylation index are more susceptible to arsenic related disease, these results suggest that GSTT1 null individuals may be more susceptible to arsenic-related toxicity. Arsenic 222-229 glutathione S-transferase theta 1 Homo sapiens 172-177 23845145-3 2013 We aimed to examine whether gene polymorphisms in GSTM1, GSTT1 and TPMT, combined with various clinical parameters, can predict thiopurine induced SAE. 2-mercaptopyrazine 128-138 glutathione S-transferase theta 1 Homo sapiens 57-62 23845145-3 2013 We aimed to examine whether gene polymorphisms in GSTM1, GSTT1 and TPMT, combined with various clinical parameters, can predict thiopurine induced SAE. SELENAZOLE-4-CARBOXYAMIDE-ADENINE DINUCLEOTIDE 147-150 glutathione S-transferase theta 1 Homo sapiens 57-62 23422534-7 2013 Two weeks on the high fruit-juice and vegetable diet increased GST and GR activities in the GSTM1+/GSTT1+ group (P < 0.05 compared with baseline or GSTM1-/GSTT1- group), although no effects were observed on GST and GR activities in GSTM1-/GSTT1- participants. juice 28-33 glutathione S-transferase theta 1 Homo sapiens 99-104 23422534-7 2013 Two weeks on the high fruit-juice and vegetable diet increased GST and GR activities in the GSTM1+/GSTT1+ group (P < 0.05 compared with baseline or GSTM1-/GSTT1- group), although no effects were observed on GST and GR activities in GSTM1-/GSTT1- participants. juice 28-33 glutathione S-transferase theta 1 Homo sapiens 158-163 23422534-7 2013 Two weeks on the high fruit-juice and vegetable diet increased GST and GR activities in the GSTM1+/GSTT1+ group (P < 0.05 compared with baseline or GSTM1-/GSTT1- group), although no effects were observed on GST and GR activities in GSTM1-/GSTT1- participants. juice 28-33 glutathione S-transferase theta 1 Homo sapiens 158-163 23506349-8 2013 Gstm1- and Gstt1-null mice have potential as human models, since null genotypes of GSTM1/Gstm1 and GSTT1/Gstt1 decreased hepatic GST activities toward p-nitrobenzyl chloride and dichloromethane, respectively, in both humans and mice. 4-nitrobenzyl chloride 151-173 glutathione S-transferase theta 1 Homo sapiens 99-104 23506349-8 2013 Gstm1- and Gstt1-null mice have potential as human models, since null genotypes of GSTM1/Gstm1 and GSTT1/Gstt1 decreased hepatic GST activities toward p-nitrobenzyl chloride and dichloromethane, respectively, in both humans and mice. 4-nitrobenzyl chloride 151-173 glutathione S-transferase theta 1 Homo sapiens 105-110 23506349-8 2013 Gstm1- and Gstt1-null mice have potential as human models, since null genotypes of GSTM1/Gstm1 and GSTT1/Gstt1 decreased hepatic GST activities toward p-nitrobenzyl chloride and dichloromethane, respectively, in both humans and mice. Methylene Chloride 178-193 glutathione S-transferase theta 1 Homo sapiens 99-104 23506349-8 2013 Gstm1- and Gstt1-null mice have potential as human models, since null genotypes of GSTM1/Gstm1 and GSTT1/Gstt1 decreased hepatic GST activities toward p-nitrobenzyl chloride and dichloromethane, respectively, in both humans and mice. Methylene Chloride 178-193 glutathione S-transferase theta 1 Homo sapiens 105-110 23330093-0 2012 Dependence of blood levels of HSP70 and HSP90 on genotypes of HSP70, GSTT1, and GSTM1 gene polymorphism in individuals chronically exposed to mercury. Mercury 142-149 glutathione S-transferase theta 1 Homo sapiens 69-74 23765968-0 2013 Influence of GSTM1 and GSTT1 polymorphisms on the survival rate of patients with malignant glioma under perillyl alcohol-based therapy. perillyl alcohol 104-120 glutathione S-transferase theta 1 Homo sapiens 23-28 23765968-3 2013 We made an explorative study of a Brazilian population with malignant glioma to determine whether GSTM1 and GSTT1 genetic polymorphisms influence the response to intranasal administration of perillyl alcohol and the survival rate. perillyl alcohol 191-207 glutathione S-transferase theta 1 Homo sapiens 108-113 23765968-10 2013 A significantly lower frequency of GSTT1 deletion in glioma patients compared to healthy controls indicates that GSTT1 deletion may exert a protective role against gliomagenesis, influence therapeutic response to intranasal perillyl alcohol treatment, and increase overall survival, especially considering tumor topography. perillyl alcohol 224-240 glutathione S-transferase theta 1 Homo sapiens 35-40 23765968-10 2013 A significantly lower frequency of GSTT1 deletion in glioma patients compared to healthy controls indicates that GSTT1 deletion may exert a protective role against gliomagenesis, influence therapeutic response to intranasal perillyl alcohol treatment, and increase overall survival, especially considering tumor topography. perillyl alcohol 224-240 glutathione S-transferase theta 1 Homo sapiens 113-118 24083736-4 2013 The homozygous null deletion of phase II metabolic gene GSTT1 that abolishes its xenobiotic- detoxifying ability may be associated with carboplatin toxicity. Carboplatin 136-147 glutathione S-transferase theta 1 Homo sapiens 56-61 24240585-10 2013 Multiple regression analysis revealed a significant interaction between beta-HCH and GSTM1<formula>^{-}</formula> genotype (p< 0.05) as well as in beta-HCH and GSTT1<formula>^{-}</formula> genotype (p< 0.05) respectively. alpha-hexachlorocyclohexane 72-80 glutathione S-transferase theta 1 Homo sapiens 175-180 23287989-3 2013 Glutathione has an important role in the defence system, catalysed by glutathione S-transferase (GST), including two non-enzyme producing polymorphisms (GSTM1-null and GSTT1-null). Glutathione 0-11 glutathione S-transferase theta 1 Homo sapiens 168-173 23099118-8 2013 Moreover, increased post-treatment ALT, AST and total bilirubin were associated with GSTM1*1/GSTT1*1 genotypes (p<0.05). Bilirubin 54-63 glutathione S-transferase theta 1 Homo sapiens 93-98 24266295-6 2013 Enzyme CYP P450 1A1, which is encoded by the CYP1A1 gene, is vital in the monooxygenation of lipofilic substrates, while GSTM1 and GSTT1 are the most abundant isophorms that conjugate and neutralize oxygen products. lipofilic 93-102 glutathione S-transferase theta 1 Homo sapiens 131-136 24266295-6 2013 Enzyme CYP P450 1A1, which is encoded by the CYP1A1 gene, is vital in the monooxygenation of lipofilic substrates, while GSTM1 and GSTT1 are the most abundant isophorms that conjugate and neutralize oxygen products. Oxygen 78-84 glutathione S-transferase theta 1 Homo sapiens 131-136 22954400-8 2012 GSTT1 was up to ten times more active than CYP 2E1 in both cell lines, indicating that potential lung damage is due to formation of pro-carcinogens such as formaldehyde. Formaldehyde 156-168 glutathione S-transferase theta 1 Homo sapiens 0-5 23259321-8 2012 Elevated 8-oxodG levels were observed in subjects with CYP1A1 CAC haplotype and GSTT1 null variant. 8-ohdg 9-16 glutathione S-transferase theta 1 Homo sapiens 80-85 23045187-12 2012 In men with functional GSTT1, the odds ratio (OR) for association of PD with paraquat use was 1.5 (95% confidence interval [CI]: 0.6-3.6); in men with GSTT1*0, the OR was 11.1 (95% CI: 3.0-44.6; P interaction: 0.027). Paraquat 77-85 glutathione S-transferase theta 1 Homo sapiens 23-28 22841242-12 2012 Anti-GSTT1 antibody was observed more frequently albeit not significantly, among the cyclosporine versus tacrolimus patient group (P = .16). Cyclosporine 85-97 glutathione S-transferase theta 1 Homo sapiens 5-10 21300142-5 2012 Results showed that occupational exposure to benzene was negligible compared to that from smoking and confirmed the modulating effect of the genetic polymorphism of GSTT1 on the urinary excretion of SPMA, but not of t, t-MA, even at very low levels of benzene exposure. Benzene 45-52 glutathione S-transferase theta 1 Homo sapiens 165-170 21300142-5 2012 Results showed that occupational exposure to benzene was negligible compared to that from smoking and confirmed the modulating effect of the genetic polymorphism of GSTT1 on the urinary excretion of SPMA, but not of t, t-MA, even at very low levels of benzene exposure. Benzene 252-259 glutathione S-transferase theta 1 Homo sapiens 165-170 22052985-4 2012 Specifically, GSTM1 and GSTT1 genes are polymorphically deleted, the polymorphism GSTP1 c.313A>G (rs1695) determines the amino acid substitution Ile105Val, where the Val-containing enzyme has reduced activity. Valine 154-157 glutathione S-transferase theta 1 Homo sapiens 24-29 22052985-4 2012 Specifically, GSTM1 and GSTT1 genes are polymorphically deleted, the polymorphism GSTP1 c.313A>G (rs1695) determines the amino acid substitution Ile105Val, where the Val-containing enzyme has reduced activity. ile105val 148-157 glutathione S-transferase theta 1 Homo sapiens 24-29 22841242-12 2012 Anti-GSTT1 antibody was observed more frequently albeit not significantly, among the cyclosporine versus tacrolimus patient group (P = .16). Tacrolimus 105-115 glutathione S-transferase theta 1 Homo sapiens 5-10 22445468-0 2012 GSTT1 gene abnormality in minimal change nephrotic syndrome with elevated serum immunoglobulin E. INTRODUCTION: Imbalance between T-helper 1 (Th1) and 2 (Th2) lymphocytes and effects of reactive oxygen species (ROS) upon glomerular capillary walls have been implicated in minimal change nephrotic syndrome (MCNS). Reactive Oxygen Species 186-209 glutathione S-transferase theta 1 Homo sapiens 0-5 22251241-1 2012 BACKGROUND: Glutathione S-transferases (GSTs) GSTM1, GSTT1 and GSTP1 are multifunctional enzymes involved in the detoxification of a wide range of reactive oxygen species produced during melanin synthesis and oxidative stress processes. Reactive Oxygen Species 147-170 glutathione S-transferase theta 1 Homo sapiens 53-58 22251241-1 2012 BACKGROUND: Glutathione S-transferases (GSTs) GSTM1, GSTT1 and GSTP1 are multifunctional enzymes involved in the detoxification of a wide range of reactive oxygen species produced during melanin synthesis and oxidative stress processes. Melanins 187-194 glutathione S-transferase theta 1 Homo sapiens 53-58 22300440-6 2012 Also, CYP19A1 arginine allele in homozygosity or heterozygosity (TC/CC) was associated with a significant increased risk for breast cancer when associated to GSTM1 null genotype (OR=6.158; 95% CI=2.676-14.171; p<0.001) and GSTT1 null genotype (OR=4.870; 95% CI=2.216-10.700; p<0.001). Arginine 14-22 glutathione S-transferase theta 1 Homo sapiens 226-231 21557334-7 2012 Flavonols and flavanols (EGC in particular) were associated with a reduced risk of breast cancer among those null for GSTM1 and GSTT1, with a P-value of 0.04 for the interaction between EGC and GSTM1 polymorphism. Flavonols 0-9 glutathione S-transferase theta 1 Homo sapiens 128-133 21557334-7 2012 Flavonols and flavanols (EGC in particular) were associated with a reduced risk of breast cancer among those null for GSTM1 and GSTT1, with a P-value of 0.04 for the interaction between EGC and GSTM1 polymorphism. flavanols 14-23 glutathione S-transferase theta 1 Homo sapiens 128-133 21557334-7 2012 Flavonols and flavanols (EGC in particular) were associated with a reduced risk of breast cancer among those null for GSTM1 and GSTT1, with a P-value of 0.04 for the interaction between EGC and GSTM1 polymorphism. gallocatechol 25-28 glutathione S-transferase theta 1 Homo sapiens 128-133 21557334-8 2012 In contrast, among women possessing both GSTM1 and GSTT1, breast cancer risk increased with levels of flavonols, particularly kaempferol. Flavonols 102-111 glutathione S-transferase theta 1 Homo sapiens 51-56 21557334-8 2012 In contrast, among women possessing both GSTM1 and GSTT1, breast cancer risk increased with levels of flavonols, particularly kaempferol. kaempferol 126-136 glutathione S-transferase theta 1 Homo sapiens 51-56 22445468-0 2012 GSTT1 gene abnormality in minimal change nephrotic syndrome with elevated serum immunoglobulin E. INTRODUCTION: Imbalance between T-helper 1 (Th1) and 2 (Th2) lymphocytes and effects of reactive oxygen species (ROS) upon glomerular capillary walls have been implicated in minimal change nephrotic syndrome (MCNS). Reactive Oxygen Species 211-214 glutathione S-transferase theta 1 Homo sapiens 0-5 22445468-1 2012 METHODS: By polymerase chain reaction and comparative genomic hybridization, we evaluated mutations of the GSTT1 gene (GSTT1), a member of the glutathione S-transferase (GST) supergene family associated with both protection of cells from ROS and control of allergic reactions and serum immunoglobulin (Ig) E. Reactive Oxygen Species 238-241 glutathione S-transferase theta 1 Homo sapiens 107-112 22445468-1 2012 METHODS: By polymerase chain reaction and comparative genomic hybridization, we evaluated mutations of the GSTT1 gene (GSTT1), a member of the glutathione S-transferase (GST) supergene family associated with both protection of cells from ROS and control of allergic reactions and serum immunoglobulin (Ig) E. Reactive Oxygen Species 238-241 glutathione S-transferase theta 1 Homo sapiens 119-124 22310945-9 2012 When interaction between GSTM1/GSTT1 genes polymorphism-OCPs levels and birth weight (gene-environment interaction) was ascertained, a significant association was seen between beta-HCH and GSTM1- genotype with reduction in birth weight of 213g. beta-hexachlorocyclohexane 176-184 glutathione S-transferase theta 1 Homo sapiens 31-36 22153878-6 2012 Furthermore the genetic polymorphism of glutathione-S-transferase (GST) may be related to health effects of benzene exposure, in fact both genotype T1 (GSTT1) and M1 (GSTM1) are involved in the detoxification of benzene oxide. Benzene 108-115 glutathione S-transferase theta 1 Homo sapiens 152-157 22153878-6 2012 Furthermore the genetic polymorphism of glutathione-S-transferase (GST) may be related to health effects of benzene exposure, in fact both genotype T1 (GSTT1) and M1 (GSTM1) are involved in the detoxification of benzene oxide. benzene oxide 212-225 glutathione S-transferase theta 1 Homo sapiens 152-157 22537952-1 2012 OBJECTIVE: To study the relationship between glutathione S-transferase genes GSTT1 and GSTM1 polymorphisms and the susceptibility to infectious mononucleosis (IM) and acute lymphocytic leukemia (ALL) in children. Glutathione 45-56 glutathione S-transferase theta 1 Homo sapiens 77-82 22170331-3 2012 In addition, in liver with GSTT1/Gstt1-null genotype, GST activity toward dichloromethane (DCM) was significantly decreased in both humans and mice. Methylene Chloride 74-89 glutathione S-transferase theta 1 Homo sapiens 27-32 21896242-8 2012 There was correlation between plasma T-AOC and consumption of F&V in the GSTM1- or GSTT1+ subjects. Adenosine Monophosphate 64-67 glutathione S-transferase theta 1 Homo sapiens 87-92 21896242-12 2012 The erythrocyte GST activity was more sensitive to consumption of F&V in the individuals with the GSTM1-/GSTT1+ genotype. f& 66-71 glutathione S-transferase theta 1 Homo sapiens 109-114 22170331-3 2012 In addition, in liver with GSTT1/Gstt1-null genotype, GST activity toward dichloromethane (DCM) was significantly decreased in both humans and mice. Methylene Chloride 74-89 glutathione S-transferase theta 1 Homo sapiens 33-38 22938433-2 2012 Thie present meta-analysis aimed to quantify the strength of the association between GSTT1 null genotype and risk of prostate cancer. thie 0-4 glutathione S-transferase theta 1 Homo sapiens 85-90 22116675-5 2012 RESULTS: Among the six candidate polymorphisms, only the distribution frequency of GSTT1 null genotype was significantly higher among AAN cases compared with controls (P = 0.041, 62.6% vs. 48.7%) and was associated with a 1.7-fold increased risk (OR = 1.728, 95%CI: 1.013-2.948, P = 0.045) of developing AAN, after adjustment for age and gender. 4-nitrophenylacetic acid 134-137 glutathione S-transferase theta 1 Homo sapiens 83-88 22116675-5 2012 RESULTS: Among the six candidate polymorphisms, only the distribution frequency of GSTT1 null genotype was significantly higher among AAN cases compared with controls (P = 0.041, 62.6% vs. 48.7%) and was associated with a 1.7-fold increased risk (OR = 1.728, 95%CI: 1.013-2.948, P = 0.045) of developing AAN, after adjustment for age and gender. 4-nitrophenylacetic acid 304-307 glutathione S-transferase theta 1 Homo sapiens 83-88 22136492-3 2012 In addition, GST-T1 and GST-M1 null genotypes have been shown to be responsible for interindividual variations in the metabolism of arsenic, a known human carcinogen. Arsenic 132-139 glutathione S-transferase theta 1 Homo sapiens 13-30 21914835-1 2011 Null mutation of glutathione transferase (GST) M1 and GSTT1 was reported to correlate statistically with an abnormal increase in the plasma levels of alanine aminotransferase or aspartate aminotransferase caused by troglitazone in diabetic patients (Clin Pharmacol Ther, 73:435-455, 2003). Troglitazone 215-227 glutathione S-transferase theta 1 Homo sapiens 54-59 21997310-3 2012 Polymorphisms of the GSTM1 and GSTT1 genes may affect ligandin functions that are important in bilirubin transportation. Bilirubin 95-104 glutathione S-transferase theta 1 Homo sapiens 31-36 22207314-5 2011 An inhibitor of MK2, a downstream regulator of p38, also diminished H(2)O(2)-induced GSTT1 upregulation. )o(2) 71-76 glutathione S-transferase theta 1 Homo sapiens 85-90 22175791-0 2012 The role of the glutathione S-transferase genes GSTT1, GSTM1, and GSTP1 in acetaminophen-poisoned patients. Glutathione 16-27 glutathione S-transferase theta 1 Homo sapiens 48-53 22175791-0 2012 The role of the glutathione S-transferase genes GSTT1, GSTM1, and GSTP1 in acetaminophen-poisoned patients. Acetaminophen 75-88 glutathione S-transferase theta 1 Homo sapiens 48-53 22291700-8 2012 GSTP1/GSTT1 and GSTP1/GSTM1 combinations showed significantly associated with increase in blood Cd levels. Cadmium 96-98 glutathione S-transferase theta 1 Homo sapiens 6-11 22291700-9 2012 This study indicated that polymorphisms of GSTP1 combined with GSTT1 and/or GSTM1 deletion are likely to influence on individual susceptibility to cadmium toxicity. Cadmium 147-154 glutathione S-transferase theta 1 Homo sapiens 63-68 22724567-8 2012 RESULTS: We have demonstrated association of early onset of cisplatin induced hearing impairment with absence of null allele of GSTT1 (p = 0.009). Cisplatin 60-69 glutathione S-transferase theta 1 Homo sapiens 128-133 22724567-10 2012 CONCLUSION: Early onset of cisplatin induced hearing impairment is more probable in persons with two functional alleles of GSTT1 gene. Cisplatin 27-36 glutathione S-transferase theta 1 Homo sapiens 123-128 23049400-3 2012 OBJECTIVE: The aim of this study was to estimate the frequency of the GSTM1 and GSTT1 genotypes in sickle cell disease patients and their effect on iron status. Iron 148-152 glutathione S-transferase theta 1 Homo sapiens 80-85 21914835-2 2011 This clinical evidence leads to the hypothesis that GSH conjugation catalyzed by GSTT1 and GSTM1 has a role in the elimination of reactive metabolites of troglitazone. Glutathione 52-55 glutathione S-transferase theta 1 Homo sapiens 81-86 21914835-2 2011 This clinical evidence leads to the hypothesis that GSH conjugation catalyzed by GSTT1 and GSTM1 has a role in the elimination of reactive metabolites of troglitazone. Troglitazone 154-166 glutathione S-transferase theta 1 Homo sapiens 81-86 21813807-0 2011 Serum vitamin C and other biomarkers differ by genotype of phase 2 enzyme genes GSTM1 and GSTT1. Ascorbic Acid 6-15 glutathione S-transferase theta 1 Homo sapiens 90-95 21961652-3 2011 The NKA, post-exposure breath naphthalene, and male gender were associated with an increase, while CYP2E1*6 DD and GSTT1-plus (++/+-) genotypes were associated with a decrease in urine naphthalene level (p < 0.0001). naphthalene 185-196 glutathione S-transferase theta 1 Homo sapiens 115-120 22058002-9 2011 Additionally, patients with the GSTT1-null genotype had higher levels of triglycerides and very low-density lipoprotein cholesterol compared to those with the GSTT1-present genotype. Triglycerides 73-86 glutathione S-transferase theta 1 Homo sapiens 32-37 21781954-1 2011 Glutathione transferase T1-1 catalyses detoxication and bioactivation processes in which glutathione conjugates are formed from endogenous and xenobiotic substrates, including alkylating agents and halogenated alkanes. Glutathione 89-100 glutathione S-transferase theta 1 Homo sapiens 0-28 21781954-1 2011 Glutathione transferase T1-1 catalyses detoxication and bioactivation processes in which glutathione conjugates are formed from endogenous and xenobiotic substrates, including alkylating agents and halogenated alkanes. Alkanes 210-217 glutathione S-transferase theta 1 Homo sapiens 0-28 21504230-11 2011 In the range of MC tested concentrations used (0.25-50 muM) GSTT1-1 and A1-1 showed a typical saturation curve with similar affinity for MC-LR ( 80 muM; k(cat) values 0.18 and 0.10 min(-1), respectively), A3-3 and M1-1 were linear, whereas GSTP1-1 showed a temperature-dependent sigmoidal allosteric curve with a k(cat) = 0.11 min(-1). Microcystins 16-18 glutathione S-transferase theta 1 Homo sapiens 60-67 21435719-4 2011 Finally, polymorphism in GSTT1 (rs4630) was associated with a lower frequency of thalidomide-induced peripheral neuropathy (p=0.04). Thalidomide 81-92 glutathione S-transferase theta 1 Homo sapiens 25-30 21798077-7 2011 Among the subjects with cumulative arsenic exposure (CAE) >= 20 mg/L*year, the GSTT1 null genotype conferred a significantly increased cancer risk (RR, 3.25, 95% CI, 1.20-8.80). Arsenic 35-42 glutathione S-transferase theta 1 Homo sapiens 82-87 21798077-8 2011 The gene-environment interaction between the GSTT1 and high arsenic exposure with respect to cancer risk was statistically significant (multiplicative model, p = 0.0151) and etiologic fraction was as high as 0.86 (95% CI, 0.51-1.22). Arsenic 60-67 glutathione S-transferase theta 1 Homo sapiens 45-50 21798077-12 2011 The genetic effects of GSTT1 and GSTO1 on arsenic-induced urothelial carcinogenesis are largely confined to very high exposure level. Arsenic 42-49 glutathione S-transferase theta 1 Homo sapiens 23-28 21152927-7 2011 Vitamin C levels varied according to GSTM1 polymorphism in the whole group (p < 0.05), in all reference subjects (p < 0.05), in the asbestos factory reference group (p < 0.05), and according to GSTT1 polymorphism in reference group of the rock wool plant (p < 0.05). Ascorbic Acid 0-9 glutathione S-transferase theta 1 Homo sapiens 203-208 21511944-1 2011 The double null mutation of glutathione transferase, GSTM1 and GSTT1, is reported to influence troglitazone-associated abnormal increases of alanine aminotransferase and aspartate aminotransferase. Troglitazone 95-107 glutathione S-transferase theta 1 Homo sapiens 63-68 21511944-6 2011 However, the CBLs of troglitazone in GSTM1/GSTT1 wild-type hepatocytes were unexpectedly higher than those in null hepatocytes. Troglitazone 21-33 glutathione S-transferase theta 1 Homo sapiens 43-48 21511944-7 2011 Although this discrepancy has not been fully explained, the GSTM1 and GSTT1 null mutations increased the cytotoxicity of troglitazone, independent of CYP3A or CYP2C8 activities. Troglitazone 121-133 glutathione S-transferase theta 1 Homo sapiens 70-75 21425430-3 2011 In our patients, DNIH was always preceded by the production of donor-specific antibodies against the glutathione S-transferase T1 (GSTT1) enzyme because of a genetic mismatch in which the donors carried the wild-type gene and the recipients displayed the null genotype. dnih 17-21 glutathione S-transferase theta 1 Homo sapiens 101-129 21425430-3 2011 In our patients, DNIH was always preceded by the production of donor-specific antibodies against the glutathione S-transferase T1 (GSTT1) enzyme because of a genetic mismatch in which the donors carried the wild-type gene and the recipients displayed the null genotype. dnih 17-21 glutathione S-transferase theta 1 Homo sapiens 131-136 21425430-4 2011 Complement component 4d (C4d) immunopositivity in 12 paraffin-embedded liver biopsy samples from 8 patients diagnosed with DNIH associated with anti-GSTT1 antibodies was retrospectively evaluated. dnih 123-127 glutathione S-transferase theta 1 Homo sapiens 149-154 21504230-11 2011 In the range of MC tested concentrations used (0.25-50 muM) GSTT1-1 and A1-1 showed a typical saturation curve with similar affinity for MC-LR ( 80 muM; k(cat) values 0.18 and 0.10 min(-1), respectively), A3-3 and M1-1 were linear, whereas GSTP1-1 showed a temperature-dependent sigmoidal allosteric curve with a k(cat) = 0.11 min(-1). cyanoginosin LR 137-142 glutathione S-transferase theta 1 Homo sapiens 60-67 22874804-4 2011 The analysis of the gene-gene interaction between GSTs indicated that the magnitude of the association was greater for the combined AG/GSTT1 & GSTM1 genotypes (OR = 2.51; 95% CI: 1.13-5.63, P = 0.02). Adenosine Monophosphate 142-145 glutathione S-transferase theta 1 Homo sapiens 135-140 21093063-3 2011 We found that the combination of the absence of GSTM1 gene with the of the GSTM1 gene with the polymorphism GSTA1*B/*B, and the presence of the GSTT1 gene, represents a risk factor for schizophrenia, indicating that the combination of different GST polymorphisms has a role in the predisposition to schizophrenia, probably affecting the capacity of the cell to detoxify the oxidized metabolites of catecholamines. Catecholamines 398-412 glutathione S-transferase theta 1 Homo sapiens 144-149 21178300-2 2011 Statistically, double null genotype of glutathione S-transferase isoforms, GSTT1 and GSTM1, was a risk factor, indicating a low activity of the susceptible patients in scavenging chemically reactive metabolites. Glutathione 39-50 glutathione S-transferase theta 1 Homo sapiens 75-80 21431478-2 2011 The aim of this study was to establish whether there is an association between the polymorphism of GSTM1 and GSTT1 and response to NACT. nact 131-135 glutathione S-transferase theta 1 Homo sapiens 109-114 21234761-2 2011 We evaluated the influence of common polymorphisms related to DNA repair or xenobiotic pathway (XRCC1, GSTP1, GSTT1, and GSTM1) on the individual susceptibility to CIHM status in the non-neoplastic rectal mucosa in UC patients. cihm 164-168 glutathione S-transferase theta 1 Homo sapiens 110-115 21352813-14 2011 Although the GSTM1-null polymorphism showed no correlation with lipid profiles among T2DM and T2DM with CAD patients, GSTT1-null polymorphism attained a statistical significance for the level of LDL (127+-28.20 vs. 134+-29.10; P=0.039) and triglycerides in T2DM with CAD patients (182.10+-21.10 vs. 191.20+-24.10; P=0.018). Triglycerides 240-253 glutathione S-transferase theta 1 Homo sapiens 118-123 20488846-0 2011 Modification of the relationship between urinary 8-OHdG and hippuric acid concentration by GSTM1, GSTT1, and ALDH2 genotypes. 8-ohdg 49-55 glutathione S-transferase theta 1 Homo sapiens 98-103 20488846-9 2011 This study shows that the relationship between urinary HA and 8-OHdG concentration is modified by genetic polymorphisms of some metabolizing enzymes such as GSTM1, GSTT1, and ALDH2. 8-ohdg 62-68 glutathione S-transferase theta 1 Homo sapiens 164-169 21097530-7 2011 In conclusion, polymorphisms of GSTT1, EPHX1, MTHFR, MTR and NAT2 differentially affect the frequency of CTAs, CSAs and CTGs, showing interaction with smoking and age. csas 111-115 glutathione S-transferase theta 1 Homo sapiens 32-37 21097530-7 2011 In conclusion, polymorphisms of GSTT1, EPHX1, MTHFR, MTR and NAT2 differentially affect the frequency of CTAs, CSAs and CTGs, showing interaction with smoking and age. ctgs 120-124 glutathione S-transferase theta 1 Homo sapiens 32-37 21619788-5 2011 After controlling potential confounders, decreased excretion of urinary 1-OHP was associated with GSTP1 I105V AG + GG genotype in coke oven workers (single-gene model, P = 0.012; multi-gene model, P = 0.011) and with GSTT1 null type in the analysis including all subjects (P = 0.055 in both single-gene and multi-gene models). Oxaliplatin 72-77 glutathione S-transferase theta 1 Homo sapiens 217-222 21734345-9 2011 Combination of 113His/His EPHX1/null-GSTM1 genotypes showed a significant association with the decrease of Delta FEV1 in patients (P =0.028).In conclusion, our results suggest combined EPHX1, GSTP1, GSTM1 and GSTT1 genetic polymorphisms may play a significant role in the development of COPD, emphysema and decline of the lung function. 113his 15-21 glutathione S-transferase theta 1 Homo sapiens 209-214 20300859-0 2011 Serum testosterone in females exposed to natural sour gas with respect to polymorphisms of XRCC1, GSTM1, and GSTT1. Testosterone 6-18 glutathione S-transferase theta 1 Homo sapiens 109-114 20300859-1 2011 The present study was done to determine the modulation effect(s) of polymorphisms of XRCC1, GSTM1, and GSTT1 on concentration of serum testosterone in females exposed to natural sour gas. Testosterone 135-147 glutathione S-transferase theta 1 Homo sapiens 103-108 20300859-8 2011 Although GSTT1-null genotype had higher level of serum testosterone in comparison with the present genotype (t=2.392, df=66, P=0.023), a borderline difference between genotypes of GSTM1 for serum testosterone was observed (t=1.928, df=66, P=0.058). Testosterone 55-67 glutathione S-transferase theta 1 Homo sapiens 9-14 20300859-8 2011 Although GSTT1-null genotype had higher level of serum testosterone in comparison with the present genotype (t=2.392, df=66, P=0.023), a borderline difference between genotypes of GSTM1 for serum testosterone was observed (t=1.928, df=66, P=0.058). Testosterone 196-208 glutathione S-transferase theta 1 Homo sapiens 9-14 20300859-9 2011 Analysis of variance revealed significant difference between combination genotypes of GSTM1 and GSTT1 for serum testosterone (F=4.167; df=3, 64; P=0.009). Testosterone 112-124 glutathione S-transferase theta 1 Homo sapiens 96-101 20300859-10 2011 The Duncan post hoc test indicated that the combination genotype of "present GSTM1/null GSTT1" had significant higher level of testosterone. Testosterone 127-139 glutathione S-transferase theta 1 Homo sapiens 88-93 20300859-12 2011 The polymorphisms of GSTM1 and GSTT1 modulate serum testosterone concentration in young females exposed to natural sour gas. Testosterone 52-64 glutathione S-transferase theta 1 Homo sapiens 31-36 21619788-6 2011 GSTT1 and GSTP1 were interacted on the urinary concentrations of 1-OHP. Oxaliplatin 65-70 glutathione S-transferase theta 1 Homo sapiens 0-5 21619788-7 2011 CONCLUSION: Urinary 1-OHP concentrations can be modified by GSTM1, GSTT1 and GSTP1 gene polymorphisms, indicating that these genes are involved in the metabolism of polycyclic aromatic hydrocarbons. Oxaliplatin 20-25 glutathione S-transferase theta 1 Homo sapiens 67-72 21619788-7 2011 CONCLUSION: Urinary 1-OHP concentrations can be modified by GSTM1, GSTT1 and GSTP1 gene polymorphisms, indicating that these genes are involved in the metabolism of polycyclic aromatic hydrocarbons. Polycyclic Aromatic Hydrocarbons 165-197 glutathione S-transferase theta 1 Homo sapiens 67-72 21156236-0 2010 GSTT1 copy number gain is a poor predictive marker for escalated-dose imatinib treatment in chronic myeloid leukemia: genetic predictive marker found using array comparative genomic hybridization. Imatinib Mesylate 70-78 glutathione S-transferase theta 1 Homo sapiens 0-5 20837120-0 2010 Susceptibility to the cytogenetic effects of dichloromethane is related to the glutathione S-transferase theta phenotype. Methylene Chloride 45-60 glutathione S-transferase theta 1 Homo sapiens 79-110 20675267-7 2010 Among the 195 cases and 192 controls with high-risk forms of GSTT1 and GSTZ1, the ORs for quartiles 2, 3, and 4 of THMs were 1.5 (0.7-3.5), 3.4 (1.4-8.2), and 5.9 (1.8-19.0), respectively. Trihalomethanes 115-119 glutathione S-transferase theta 1 Homo sapiens 61-66 20194072-0 2010 Interaction between GSTM1/GSTT1 polymorphism and blood mercury on birth weight. Mercury 55-62 glutathione S-transferase theta 1 Homo sapiens 26-31 20042523-6 2010 The strongest inverse association was found among individuals with both the GSTM1-null and the GSTT1-null genotypes, with an adjusted odds ratio of 0.51 (95% CI: 0.27, 0.95), in a comparison of the highest with the lowest tertile of urinary isothiocyanates. Isothiocyanates 241-256 glutathione S-transferase theta 1 Homo sapiens 95-100 20042523-8 2010 CONCLUSION: This study suggests that isothiocyanate exposure may reduce the risk of colorectal cancer, and this protective effect may be modified by the GSTM1 and GSTT1 genes. isothiocyanic acid 37-51 glutathione S-transferase theta 1 Homo sapiens 163-168 20638463-3 2010 We conducted a study to determine whether N-Acetyl-cysteine (NAC) protected men against noise-induced temporary threshold shift (TTS), and whether subgroups with genetic polymorphisms of glutathione S-transferase (GST) T1 and M1 responded to NAC differently. Acetylcysteine 242-245 glutathione S-transferase theta 1 Homo sapiens 187-228 20638463-11 2010 When the participants were grouped by GST M1/T1 genotypes, the NAC effect was only significant among workers with null genotypes in both GSTM1 and GSTT1 (p = 0.004). Acetylcysteine 63-66 glutathione S-transferase theta 1 Homo sapiens 147-152 20638463-13 2010 The protective effect of NAC was more prominent in subjects with both GSTM1-null and GSTT1-null genotypes. Acetylcysteine 25-28 glutathione S-transferase theta 1 Homo sapiens 85-90 20663906-7 2010 A significant association was found among TCE-exposed subjects with at least one intact GSTT1 allele (active genotype; OR = 1.88; 95% CI, 1.06-3.33) but not among subjects with two deleted alleles (null genotype; OR = 0.93; 95% CI, 0.35-2.44; P(interaction) = 0.18). Trichloroethylene 42-45 glutathione S-transferase theta 1 Homo sapiens 88-93 19838709-5 2010 Patients who had null genotype for both the alleles, i.e., GSTT1/GSTM1 had significantly higher levels of serum iron (P = 0.007) and serum ferritin (P = 0.001) than patients with normal genotype for GST deletions. Iron 112-116 glutathione S-transferase theta 1 Homo sapiens 59-64 20042523-5 2010 RESULTS: Urinary isothiocyanate concentrations were inversely associated with colorectal cancer risk; the inverse association was statistically significant or nearly significant in the GSTM1-null (P for trend = 0.04) and the GSTT1-null (P for trend = 0.07) genotype groups. isothiocyanic acid 17-31 glutathione S-transferase theta 1 Homo sapiens 225-230 20200426-9 2010 Furthermore, GSTM4, GSTT1, and ABCC4 overexpression significantly decreased cisplatin sensitivity in lung cancer and HEK293T cell lines. Cisplatin 76-85 glutathione S-transferase theta 1 Homo sapiens 20-25 20200426-11 2010 ABCC4 polymorphisms, as well as GSTT1 copy number, may also help to predict cisplatin response, but further validation is required. Cisplatin 76-85 glutathione S-transferase theta 1 Homo sapiens 32-37 21637441-2 2009 We tested whether the null genotypes of the GSTM1 and GSTT1 genes, involved in benzene inactivation, altered the risk for NEB in southeastern Brazil. Benzene 79-86 glutathione S-transferase theta 1 Homo sapiens 54-59 20056632-9 2010 Those with deletions in both GSTM1 and GSTT1 genes combined had a significantly reduced risk with increasing glucosinolate intake (P(interaction) = 0.01). Glucosinolates 109-122 glutathione S-transferase theta 1 Homo sapiens 39-44 20056632-11 2010 This study showed that the inverse association between glucosinolate intake and prostate cancer risk was modified by NQO1 (C609T) and GSTM1 and GSTT1 deletion polymorphisms. Glucosinolates 55-68 glutathione S-transferase theta 1 Homo sapiens 144-149 20464961-0 2010 [Association of genetic polymorphism in GSTM1 and GSTT1 with the frequency of chromosomal aberrations in the uranium workers]. Uranium 109-116 glutathione S-transferase theta 1 Homo sapiens 50-55 20464961-1 2010 Association between genetic polymorphism in GSTM1 and GSTT1 and frequency of chromosomal aberrations in the workers occupationally exposed to uranium during 1-25 years in Tselinniy mining/milling complex in the North Kazakhstan was investigated. Uranium 142-149 glutathione S-transferase theta 1 Homo sapiens 54-59 19710200-6 2009 RESULTS: A gene-diet interaction on serum ascorbic acid was observed for GSTM1 (P = 0.04) and GSTT1 (P = 0.01) but not for GSTP1 (P = 0.83). Ascorbic Acid 42-55 glutathione S-transferase theta 1 Homo sapiens 94-99 19741569-10 2009 Failure, after platinum-based chemotherapy, was associated with the GSTT1positive/GSTP-AA or GSTP-GG/GSTM1-positive genotype (P = 0.019, OR: 2.168, 95% CI: 1.130-4.160). Platinum 15-23 glutathione S-transferase theta 1 Homo sapiens 68-73 19843381-4 2009 In this study, the expression of the wild and mutant type of the GSTT-1 gene of those stable transformants in cell lines and in bone marrow cells from MDS patients by reverse-transcription polymerase chain reaction (RT-PCR) was observed in the presence or absence of rapamycin. Sirolimus 267-276 glutathione S-transferase theta 1 Homo sapiens 65-71 19843381-5 2009 Significant growth inhibition by rapamycin was observed among stable transformants for the mutant GSTT-1 gene, but not wild type GSTT-1 gene, and was indicative of typical apoptosis. Sirolimus 33-42 glutathione S-transferase theta 1 Homo sapiens 98-104 19419718-7 2009 CONCLUSIONS: The data suggest that the presence of a double deletion genotypes of the GSTM1 and GSTT1 genes is associated with hypertriglyceridemia and low HDL-cholesterol levels in humans. Cholesterol 160-171 glutathione S-transferase theta 1 Homo sapiens 96-101 18642130-0 2009 Serum levels of testosterone and gonadotrophins with respect to smoking status and genetic polymorphism of GSTT1. Testosterone 16-28 glutathione S-transferase theta 1 Homo sapiens 107-112 19097986-4 2009 AZM exposure significantly decreased GSTT1 and GSTM1 mRNA and protein expression in IB3-1, restoring the levels to those observed in non-CF C38 cells, which also express lower levels of gamma-glutamyltransferase (GGT) activity than IB3-1. Azithromycin 0-3 glutathione S-transferase theta 1 Homo sapiens 37-42 19097986-5 2009 In another CF cell line, 2CFSMEo-, AZM produced 45% reduction in GSTT1 and GSTM1 mRNA levels. Azithromycin 35-38 glutathione S-transferase theta 1 Homo sapiens 65-70 19515364-11 2009 GSTP1-114/GSTT1 and GSTP1-105/GCLC combinations showed synergistic effects on hair mercury levels compared to single-gene variants. Mercury 83-90 glutathione S-transferase theta 1 Homo sapiens 10-15 19548560-0 2009 [Influence of PAHs exposure and GSTT1, GSTM1 genotypes on urinary 1-OHP as exposure biomarker]. Oxaliplatin 66-71 glutathione S-transferase theta 1 Homo sapiens 32-37 19303595-6 2009 The risk of low motility with high DDE-DDT exposure was increased in men with the GSTT1 null genotype compared to those with GSTT1 intact (odds ratio (OR)=4.19, 95% confidence interval (CI) 1.05-16.78 and OR=3.57, 1.43-8.93, respectively). Dichlorodiphenyl Dichloroethylene 35-38 glutathione S-transferase theta 1 Homo sapiens 82-87 19303595-10 2009 CONCLUSION: High DDE-DDT exposure adversely affected all 3 sperm parameters and its effects were exacerbated by the GSTT1 null polymorphism and by the CYP1A1 common alleles. Dichlorodiphenyl Dichloroethylene 17-20 glutathione S-transferase theta 1 Homo sapiens 116-121 19927646-0 2009 [Abnormal liver function associated with polymorphism of GSTT1, GSTM1 and CYP2E1 in workers exposed to N, N-dimethylformamide]. Dimethylformamide 103-125 glutathione S-transferase theta 1 Homo sapiens 57-62 19927646-1 2009 OBJECTIVE: To investigate abnormal liver function associated with polymorphism of GSTT1, GSTM1 and CYP2E1 in workers exposed to N, N-dimethylformamide. Dimethylformamide 128-150 glutathione S-transferase theta 1 Homo sapiens 82-87 19157724-1 2009 In a case-control study, association of polymorphism in glutathione-S-transferases (GSTM1, GSTT1, GSTP1), involved in detoxification of reactive oxygen species (ROS), was studied with alcoholic liver cirrhosis. Reactive Oxygen Species 136-159 glutathione S-transferase theta 1 Homo sapiens 91-96 19548560-1 2009 OBJECTIVE: To study the influence of polycyclic aromatic hydrocarbons (PAHs) exposure (ambient concentration and smoking) and GSTT1, GSTM1, genotypes on urinary 1-OHP as exposure biomarker. Oxaliplatin 161-166 glutathione S-transferase theta 1 Homo sapiens 126-131 19131562-7 2009 The ratio of GA-Hb/AA-Hb in individuals with the combined GSTM1- and GSTT1-null variants was significantly (p = 0.029) higher than those with the wild-type genotypes. glycidamide 13-15 glutathione S-transferase theta 1 Homo sapiens 69-74 19388510-3 2009 The most widely studied group were genes encoded molecules engaged in biotransformations of xenobiotics, in particular potential carcinogens, like alcohol (ADH2) and aldehyde (ALDH2) dehydrogenases, various isoenzymes of cytochrome P450 (CYP1A1, CYP2E1) and glutathione S-transferase (GSTM1, GSTT1, GSTP1). Alcohols 147-154 glutathione S-transferase theta 1 Homo sapiens 292-297 18449862-2 2009 In this study, we performed association studies between GSH-related genes (GSTM1, GSTP1, GSTO1, GSTT1, GSTT2, GPX1, and GCLM) and schizophrenia in a Japanese population. Glutathione 56-59 glutathione S-transferase theta 1 Homo sapiens 96-101 18559562-13 2008 Using SPMA for evaluating benzene exposure, the results suggest that the GSTT1 genetic polymorphism, especially in a comparison study between two populations with different GSTT1 genotype frequencies, should be considered. Benzene 26-33 glutathione S-transferase theta 1 Homo sapiens 73-78 17874314-1 2008 OBJECTIVE: In order to examine whether chronic exposure to natural sour gas containing sulfur compounds act as natural selection force on genetic polymorphisms of glutathione S-transferase M1 (GSTM1) and T1 (GSTT1), the present study was done. Sulfur 87-93 glutathione S-transferase theta 1 Homo sapiens 208-213 18186040-4 2008 There was a significant difference in GSTT1 genotype frequency between patients with METH psychosis and controls (P = 0.039, odds ratio: 1.52, 95% CI 1.03-2.24). Methamphetamine 85-89 glutathione S-transferase theta 1 Homo sapiens 38-43 18186040-5 2008 Furthermore, the frequency (66.0%) of the GSTT1 null genotype among prolonged-type METH psychotic patients with spontaneous relapse was significantly higher (P = 0.025, odds ratio: 2.43, 95% CI 1.13-5.23) than that (44.4%) of transient-type METH psychotic patients without spontaneous relapse. Methamphetamine 83-87 glutathione S-transferase theta 1 Homo sapiens 42-47 18186040-5 2008 Furthermore, the frequency (66.0%) of the GSTT1 null genotype among prolonged-type METH psychotic patients with spontaneous relapse was significantly higher (P = 0.025, odds ratio: 2.43, 95% CI 1.13-5.23) than that (44.4%) of transient-type METH psychotic patients without spontaneous relapse. Methamphetamine 241-245 glutathione S-transferase theta 1 Homo sapiens 42-47 18186040-7 2008 The present study suggests that the polymorphism of the GSTT1 gene might be a genetic risk factor of the development of METH psychosis in a Japanese population. Methamphetamine 120-124 glutathione S-transferase theta 1 Homo sapiens 56-61 18577398-0 2008 CYP1A1 Ile462Val and GSTT1 modify the effect of cord blood cotinine on neurodevelopment at 2 years of age. Cotinine 59-67 glutathione S-transferase theta 1 Homo sapiens 21-26 18577398-12 2008 It can be concluded that CYP1A1 Ile462Val and GSTT1 metabolic genes can modify the effect of cord blood cotinine on early child neurodevelopment especially for language and fine motor development. Cotinine 104-112 glutathione S-transferase theta 1 Homo sapiens 46-51 19317600-1 2008 Cytochrome 1A1 (CYP1A1), glutathione transferase M1 (GSTM1), and glutathione transferase T1 (GSTT1) catalyze the bioactivation and detoxification of a wide variety of xenobiotic compounds that are mutagenic and/or carcinogenic (e.g., polycyclic aromatic hydrocarbons). Polycyclic Aromatic Hydrocarbons 234-266 glutathione S-transferase theta 1 Homo sapiens 93-98 18641537-3 2008 In this report, the contribution of genetic polymorphisms in the glutathione-S-transferases (GST) isozymes GSTA1, GSTM1, GSTP1, and GSTT1 to the pharmacokinetics of busulfan is studied retrospectively. Busulfan 165-173 glutathione S-transferase theta 1 Homo sapiens 132-137 18839526-0 2008 [Study on the relationship between GSTM1, GSTT1 gene polymorphisms and arsenic methylation level]. Arsenic 71-78 glutathione S-transferase theta 1 Homo sapiens 42-47 18839526-1 2008 OBJECTIVE: To investigate the relationship between genetic polymorphisms of GSTT1, GSTM1 and arsenic methylation level. Arsenic 93-100 glutathione S-transferase theta 1 Homo sapiens 76-81 19252342-3 2009 In this study, to elucidate the haplotype structures of GSTT1 and GSTM1, genetic variations were identified in 194 Japanese cancer patients who received platinum-based chemotherapy. Platinum 153-161 glutathione S-transferase theta 1 Homo sapiens 56-61 19057715-2 2008 We hypothesized that genetic variability in glutathione S-transferase (GST) genes (GSTP1, GSTM1, and GSTT1) could influence the effects of prenatal exposure to p,p"-DDT. DDT 160-168 glutathione S-transferase theta 1 Homo sapiens 101-106 18594869-11 2008 Our results indicate that among children with steroid-sensitive NS, there is an association with response to IVCP therapy and combination of GSTP1 Val105 polymorphism and the null genotypes of GSTT1 and GSTM1. Steroids 46-53 glutathione S-transferase theta 1 Homo sapiens 193-198 18608187-1 2008 The aim of this work was to investigate urinary analytes and haemoglobin and albumin adducts as biomarkers of exposure to airborne styrene (Sty) and styrene-(7,8)-oxide (StyOX) and to evaluate the influence of smoking habit and genetic polymorphism of metabolic enzymes GSTM1 and GSTT1 on these biomarkers. styrene oxide 170-175 glutathione S-transferase theta 1 Homo sapiens 280-285 19103108-1 2008 OBJECTIVE: To study the potential effect of gene-environment interaction between glutathione S-transferase T1 (GSTT1) and serum organochlorines residues on the risk of breast cancer in women, in China. Hydrocarbons, Chlorinated 128-143 glutathione S-transferase theta 1 Homo sapiens 81-109 19103108-6 2008 RESULTS: After adjusting the confounding factors, results showed that interaction existed in genetic polymorphisms of GSTT1 and dichlorodiphenyltrichloroethane (DDT)/hexachlorocyclohexane (HCH) residues, with interaction indexes (gamma) value as 1.352 and 1.528. DDT 128-159 glutathione S-transferase theta 1 Homo sapiens 118-123 18559562-13 2008 Using SPMA for evaluating benzene exposure, the results suggest that the GSTT1 genetic polymorphism, especially in a comparison study between two populations with different GSTT1 genotype frequencies, should be considered. Benzene 26-33 glutathione S-transferase theta 1 Homo sapiens 173-178 17953974-0 2008 Inherited susceptibility to bleomycin-induced micronuclei: correlating polymorphisms in GSTT1, GSTM1 and DNA repair genes with mutagen sensitivity. Bleomycin 28-37 glutathione S-transferase theta 1 Homo sapiens 88-93 17880951-10 2008 The LH levels varied between the two groups; the mean serum LH was lower in the exposed versus unexposed men who possessed the GSTT1 genotype (2.82 +/- 1.32 IU/L vs. 3.20 +/- 1.17 IU/L) and higher in the exposed versus unexposed men who possessed the GSTT1-null genotype (3.52 +/- 1.37 IU/L vs. 2.76 +/- 1.07 IU/L). Luteinizing Hormone 60-62 glutathione S-transferase theta 1 Homo sapiens 127-132 17912498-4 2008 Safrole may be metabolized by hepatic sulfotransferase 1A1 (SULT1A1), or glutathione S-transferases (GSTM1, GSTT1, and GSTP1). Safrole 0-7 glutathione S-transferase theta 1 Homo sapiens 108-113 18325667-1 2008 In order to investigate the association between effects of genetic polymorphisms of GSTT1 and GSTM1 and depression score of individuals chronically exposed to natural sour gas containing sulfur compounds, the present cross-sectional study was done. Sulfur 187-193 glutathione S-transferase theta 1 Homo sapiens 84-89 18155166-0 2008 Glutathione-S-transferase (GST) M1 null genotype and combined GSTM1 and GSTT1 null genotypes are risk factors for increased serum gamma-glutamyltransferase in valproic acid-treated patients. Valproic Acid 159-172 glutathione S-transferase theta 1 Homo sapiens 72-77 18155166-7 2008 CONCLUSIONS: The GSTM1- and GSTM1-/GSTT1- genotypes may be a genetic risk factor for the increase of GGT in VPA-treated patients. Valproic Acid 108-111 glutathione S-transferase theta 1 Homo sapiens 35-40 18303971-6 2008 Genetic variations in the glutathione S-transferases GSTT1 and GSTM1 have been studied in many human populations, and association of these variations with environmentally-related cancers, drug-induced hepatotoxicity and even chronification of viral hepatitis has been shown. Glutathione 26-37 glutathione S-transferase theta 1 Homo sapiens 53-58 17975885-8 2007 GSTT1-1 hardly catalyzes the glutathione conjugation of curcumin. Glutathione 29-40 glutathione S-transferase theta 1 Homo sapiens 0-7 18035380-1 2008 Polymorphism in glutathione S-transferase (GST) genes (GSTM1, GSTT1 and GSTP1) and interaction with environmental factors such as tobacco (smoking or chewing) and alcohol on susceptibility to head and neck squamous cell carcinoma (HNSCC) was studied in a case-control study. Alcohols 163-170 glutathione S-transferase theta 1 Homo sapiens 43-46 17975885-8 2007 GSTT1-1 hardly catalyzes the glutathione conjugation of curcumin. Curcumin 56-64 glutathione S-transferase theta 1 Homo sapiens 0-7 17513527-5 2007 METHODS: In the present study, the association between ARHI and polymorphisms in genes that contribute to the defence against reactive oxygen species, including GSTT1, GSTM1 and NAT2, was tested. Reactive Oxygen Species 126-149 glutathione S-transferase theta 1 Homo sapiens 161-166 17980649-12 2007 A recent epidemiologic study found that much of the risk for bladder cancer associated with drinking water was associated with three factors: THM levels, showering/bathing/swimming (i.e., dermal/inhalation exposure), and genotype (having the GSTT1-1 gene). Water 101-106 glutathione S-transferase theta 1 Homo sapiens 242-249 17716707-11 2007 RESULTS: The following was noted: a) hair mercury concentrations are significantly increased in persons with the double deleted genotype (GSTT1-/- and GSTM1-/-) as compared to persons with the intact genotype, and b) MT1X expression is higher in persons with the intact genotype (GSTT1+/+ and GSTM1+/+). Mercury 42-49 glutathione S-transferase theta 1 Homo sapiens 138-143 17716707-11 2007 RESULTS: The following was noted: a) hair mercury concentrations are significantly increased in persons with the double deleted genotype (GSTT1-/- and GSTM1-/-) as compared to persons with the intact genotype, and b) MT1X expression is higher in persons with the intact genotype (GSTT1+/+ and GSTM1+/+). Mercury 42-49 glutathione S-transferase theta 1 Homo sapiens 280-285 17716707-12 2007 CONCLUSIONS: We conclude that the epistatic effect of the GSTT1 and the GSTM1 deletion polymorphism is a risk factor for increased susceptibility to mercury exposure. Mercury 149-156 glutathione S-transferase theta 1 Homo sapiens 58-63 17659824-2 2007 Cytochrome P450 2E1 (CYP2E1), aldehyde dehydrogenase 2 (ALDH2) and glutathione S-transferase theta 1 (GSTT1) enzymes are involved in activation and detoxification of VCM, and thus may be important determinants of interindividual susceptibility to VCM-induced liver damage, including liver cirrhosis. Vinyl Chloride 166-169 glutathione S-transferase theta 1 Homo sapiens 67-100 17659824-2 2007 Cytochrome P450 2E1 (CYP2E1), aldehyde dehydrogenase 2 (ALDH2) and glutathione S-transferase theta 1 (GSTT1) enzymes are involved in activation and detoxification of VCM, and thus may be important determinants of interindividual susceptibility to VCM-induced liver damage, including liver cirrhosis. Vinyl Chloride 166-169 glutathione S-transferase theta 1 Homo sapiens 102-107 17307802-1 2007 To test the hypothesis of interaction among genetic variants in increasing the individual risk of cancer, we have studied the cumulative effect on lung cancer risk of variants in three metabolic genes, CYP1A1, GSTM1 and GSTT1, which are involved in the metabolism of the tobacco smoke constituents and environmental contaminants, polycyclic aromatic hydrocarbons and of other lung carcinogens. Polycyclic Aromatic Hydrocarbons 330-362 glutathione S-transferase theta 1 Homo sapiens 220-225 17706336-0 2007 GSTM1 and GSTT1 polymorphism influences protection against induced oxidative DNA damage by quercetin and ascorbic acid in human lymphocytes in vitro. Quercetin 91-100 glutathione S-transferase theta 1 Homo sapiens 10-15 17706336-0 2007 GSTM1 and GSTT1 polymorphism influences protection against induced oxidative DNA damage by quercetin and ascorbic acid in human lymphocytes in vitro. Ascorbic Acid 105-118 glutathione S-transferase theta 1 Homo sapiens 10-15 17706336-2 2007 This study aims to establish the impact of genetic polymorphisms in GSTM1 and GSTT1, which encode for enzymatic antioxidative defence, on H(2)O(2)-induced oxidative DNA damage and on the effectiveness of quercetin and ascorbic acid in preventing this induced damage in human lymphocytes. Quercetin 204-213 glutathione S-transferase theta 1 Homo sapiens 78-83 17706336-2 2007 This study aims to establish the impact of genetic polymorphisms in GSTM1 and GSTT1, which encode for enzymatic antioxidative defence, on H(2)O(2)-induced oxidative DNA damage and on the effectiveness of quercetin and ascorbic acid in preventing this induced damage in human lymphocytes. Ascorbic Acid 218-231 glutathione S-transferase theta 1 Homo sapiens 78-83 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Hydrogen Peroxide 23-31 glutathione S-transferase theta 1 Homo sapiens 102-107 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Hydrogen Peroxide 23-31 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Hydrogen Peroxide 23-31 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Hydrogen Peroxide 23-31 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Quercetin 64-73 glutathione S-transferase theta 1 Homo sapiens 102-107 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Quercetin 64-73 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Quercetin 64-73 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Quercetin 64-73 glutathione S-transferase theta 1 Homo sapiens 127-132 17706336-8 2007 The protection against H(2)O(2)-induced oxidative DNA damage by quercetin was significantly higher in GSTT1 wild types than in GSTT1 variants (57% and 9% decrease, respectively; p=0.01); furthermore, GSTT1 wild types were protected against induced oxidative DNA damage by ascorbic acid pre-incubation while GSTT1 variants showed an increase of damage (16% decrease vs. 91% increase; p=0.01). Ascorbic Acid 272-285 glutathione S-transferase theta 1 Homo sapiens 102-107 17885617-7 2007 Results generally support prior associations between benzene hematotoxicity and specific gene mutations, confirm earlier evidence that GSTT1 affects production of S-phenylmercapturic acid, and provide additional evidence that genetic polymorphisms in NQO1*2, CYP2E1, and EPHX1 (rs1051740 or rs2234922) affect metabolism of benzene in the human liver. S-phenyl-N-acetylcysteine 163-187 glutathione S-transferase theta 1 Homo sapiens 135-140 17445838-9 2007 Furthermore the significant inverse correlation between 8-oxodG and B[a]P DNA adducts was confined to individuals carrying the wild type genotype for both the GSTM1 and the GSTT1 gene (separately and interacting). 8-ohdg 56-63 glutathione S-transferase theta 1 Homo sapiens 173-178 17498780-6 2007 GSTT1 present carriers had a significantly higher urinary 1-OHP level than that in null carriers in the case with AhR R554K GA/AA carriers (5.17 vs. 3.64 micromol/mol creatinine, p=0.038), as well as in the case with UGT1A1 -3263T>G TG/GG carriers (5.67 vs. 3.38 micromol/mol creatinine, p=0.001). Oxaliplatin 58-63 glutathione S-transferase theta 1 Homo sapiens 0-5 17498780-6 2007 GSTT1 present carriers had a significantly higher urinary 1-OHP level than that in null carriers in the case with AhR R554K GA/AA carriers (5.17 vs. 3.64 micromol/mol creatinine, p=0.038), as well as in the case with UGT1A1 -3263T>G TG/GG carriers (5.67 vs. 3.38 micromol/mol creatinine, p=0.001). Creatinine 167-177 glutathione S-transferase theta 1 Homo sapiens 0-5 17498780-6 2007 GSTT1 present carriers had a significantly higher urinary 1-OHP level than that in null carriers in the case with AhR R554K GA/AA carriers (5.17 vs. 3.64 micromol/mol creatinine, p=0.038), as well as in the case with UGT1A1 -3263T>G TG/GG carriers (5.67 vs. 3.38 micromol/mol creatinine, p=0.001). Thioguanine 236-238 glutathione S-transferase theta 1 Homo sapiens 0-5 17498780-6 2007 GSTT1 present carriers had a significantly higher urinary 1-OHP level than that in null carriers in the case with AhR R554K GA/AA carriers (5.17 vs. 3.64 micromol/mol creatinine, p=0.038), as well as in the case with UGT1A1 -3263T>G TG/GG carriers (5.67 vs. 3.38 micromol/mol creatinine, p=0.001). Creatinine 279-289 glutathione S-transferase theta 1 Homo sapiens 0-5 17498780-7 2007 These results showed that AhR, UGT1A1, GSTP1 and GSTT1 polymorphisms were associated with urinary 1-OHP concentrations in Chinese coke oven workers. Oxaliplatin 98-103 glutathione S-transferase theta 1 Homo sapiens 49-54 17310693-9 2007 Bladder cancer risk from THM exposure (all routes combined) was greatest among those with the GSTT1-1 gene. Trihalomethanes 25-28 glutathione S-transferase theta 1 Homo sapiens 94-101 17450230-9 2007 We also saw minor effects of other polymorphisms in the multivariate regression analysis with effect modification for the deletion genotypes for GSTM1 (affecting %MMA) and GSTT1 (affecting %MMA and %DMA). dma 199-202 glutathione S-transferase theta 1 Homo sapiens 172-177 17431481-7 2007 We observed significant interaction on the multiplicative scale between GSTT1 wildtype and secondary methylation ratio [dimethylarsinic acid/MMA; likelihood ratio test (LRT), p = 0.01]. Cacodylic Acid 120-140 glutathione S-transferase theta 1 Homo sapiens 72-77 17431481-7 2007 We observed significant interaction on the multiplicative scale between GSTT1 wildtype and secondary methylation ratio [dimethylarsinic acid/MMA; likelihood ratio test (LRT), p = 0.01]. monomethylarsonic acid 141-144 glutathione S-transferase theta 1 Homo sapiens 72-77 17431481-10 2007 The interaction between GSTT1 wildtype and secondary methylation ratio modifies risk of skin lesions among arsenic-exposed individuals. Arsenic 107-114 glutathione S-transferase theta 1 Homo sapiens 24-29 17330842-0 2007 Role of GSTT1 deletion in DNA oxidative damage by exposure to polycyclic aromatic hydrocarbons in humans. Polycyclic Aromatic Hydrocarbons 62-94 glutathione S-transferase theta 1 Homo sapiens 8-13 17330842-3 2007 The GSTT1 homozygous deletion variant was associated with a significant protective effect for exposure to total PAHs and to eight specific PAHs, although the magnitude and significance of the effect varied among these compounds. Polycyclic Aromatic Hydrocarbons 112-116 glutathione S-transferase theta 1 Homo sapiens 4-9 17330842-3 2007 The GSTT1 homozygous deletion variant was associated with a significant protective effect for exposure to total PAHs and to eight specific PAHs, although the magnitude and significance of the effect varied among these compounds. Polycyclic Aromatic Hydrocarbons 139-143 glutathione S-transferase theta 1 Homo sapiens 4-9 17330842-4 2007 Categorical sensitivity analysis was used to determine that the frequency of the GSTT1 deletion was significantly higher in people who proved to be more resistant to the DNA damaging effects of PAH exposure than in people who were the most sensitive. Polycyclic Aromatic Hydrocarbons 194-197 glutathione S-transferase theta 1 Homo sapiens 81-86 17330842-6 2007 The mechanism for this effect might be related to specific PAH substrate specificities, or could be related to other functions of GSTT1 gene in oxidative stress induced damage pathways. Polycyclic Aromatic Hydrocarbons 59-62 glutathione S-transferase theta 1 Homo sapiens 130-135 17465707-1 2007 UNLABELLED: The aim of this study is to verify whether the combination of glutathione S-transferase (GST) M1 null and GSTT1 null genotypes, which is a candidate genetic risk factor for troglitazone-induced liver failure, is common to that for the carbamazepine-induced mild hepatotoxicity. Troglitazone 185-197 glutathione S-transferase theta 1 Homo sapiens 118-123 17465707-1 2007 UNLABELLED: The aim of this study is to verify whether the combination of glutathione S-transferase (GST) M1 null and GSTT1 null genotypes, which is a candidate genetic risk factor for troglitazone-induced liver failure, is common to that for the carbamazepine-induced mild hepatotoxicity. Carbamazepine 247-260 glutathione S-transferase theta 1 Homo sapiens 118-123 17339179-10 2007 and del{GSTT1} polymorphisms showed a higher risk than females of developing AL. Aluminum 77-79 glutathione S-transferase theta 1 Homo sapiens 8-13 17182005-1 2007 The aim of this study is to investigate GSTM1, GSTT1 and MTHFR genetic polymorphisms and its relation with total plasma glutathione (tGSH) levels in hypertension. Glutathione 120-131 glutathione S-transferase theta 1 Homo sapiens 47-52 16406813-8 2007 CONCLUSION: Our study shows that exposure to urban traffic, dietary habits, and the nonnull GSTT1 genotype may contribute to interindividual variation in background levels of 1-OHP urinary excretion in subjects without occupational exposure to PAHs. Oxaliplatin 175-180 glutathione S-transferase theta 1 Homo sapiens 92-97 16406813-8 2007 CONCLUSION: Our study shows that exposure to urban traffic, dietary habits, and the nonnull GSTT1 genotype may contribute to interindividual variation in background levels of 1-OHP urinary excretion in subjects without occupational exposure to PAHs. Polycyclic Aromatic Hydrocarbons 244-248 glutathione S-transferase theta 1 Homo sapiens 92-97 17119257-8 2006 Variation in multiple benzene metabolizing genes may be associated with risk of benzene hematotoxicity, including CYP2E1, MPO, NQO1, and GSTT1. Benzene 22-29 glutathione S-transferase theta 1 Homo sapiens 137-142 18333132-9 2007 RESULTS: Statistical analysis of the microarray data showed that four genes were differentially expressed in gemcitabine-sensitive cancers: microsomal glutathione S-transferase 1 (GSTT1), topoisomerase II alpha (TOP2A), caspase 3, and ATP-binding cassette and subfamily C member 2 (ABCC2). gemcitabine 109-120 glutathione S-transferase theta 1 Homo sapiens 180-185 17078101-4 2006 Analysis of the effect of pairs of genes showed that for a fixed GSTT1 genotype, the SCE level increased with an increasing number of Tyr alleles in EPHX codon 113. Tyrosine 134-137 glutathione S-transferase theta 1 Homo sapiens 65-70 17078101-5 2006 We found that among GSTT1(+) individuals the DEB-induced SCE level was significantly lower when the EPHX 139 codon was His/Arg rather than His/His. Histidine 119-122 glutathione S-transferase theta 1 Homo sapiens 20-25 17078101-5 2006 We found that among GSTT1(+) individuals the DEB-induced SCE level was significantly lower when the EPHX 139 codon was His/Arg rather than His/His. Arginine 123-126 glutathione S-transferase theta 1 Homo sapiens 20-25 17078101-5 2006 We found that among GSTT1(+) individuals the DEB-induced SCE level was significantly lower when the EPHX 139 codon was His/Arg rather than His/His. Histidine 139-142 glutathione S-transferase theta 1 Homo sapiens 20-25 17078101-5 2006 We found that among GSTT1(+) individuals the DEB-induced SCE level was significantly lower when the EPHX 139 codon was His/Arg rather than His/His. Histidine 139-142 glutathione S-transferase theta 1 Homo sapiens 20-25 17011574-2 2006 For example, the human theta 1-1 enzyme (hGSTT1-1) is 440-fold less efficient than the rat theta 2-2 enzyme (rGSTT2-2) with the fluorogenic substrate 7-amino-4-chloromethyl coumarin (CMAC). 7-amino-4-chloromethylcoumarin 150-181 glutathione S-transferase theta 1 Homo sapiens 41-49 17011574-2 2006 For example, the human theta 1-1 enzyme (hGSTT1-1) is 440-fold less efficient than the rat theta 2-2 enzyme (rGSTT2-2) with the fluorogenic substrate 7-amino-4-chloromethyl coumarin (CMAC). 7-amino-4-chloromethylcoumarin 183-187 glutathione S-transferase theta 1 Homo sapiens 41-49 17011574-3 2006 Large libraries of hGSTT1-1 constructed by error-prone PCR, DNA shuffling, or saturation mutagenesis were screened for improved catalytic activity towards CMAC in a quantitative fashion using flow cytometry. 7-amino-4-chloromethylcoumarin 155-159 glutathione S-transferase theta 1 Homo sapiens 19-27 16874663-3 2006 Typical substrates for GSTT1 are industrial compounds, such as dichloromethane and ethylene oxide. Methylene Chloride 63-78 glutathione S-transferase theta 1 Homo sapiens 23-28 16874663-3 2006 Typical substrates for GSTT1 are industrial compounds, such as dichloromethane and ethylene oxide. Ethylene Oxide 83-97 glutathione S-transferase theta 1 Homo sapiens 23-28 16874663-4 2006 It has been shown that also chemotherapeutic drugs such as BCNU [i.e. 1,3-bis(2-chloroethyl)-1-nitrosourea] are efficiently inactivated by GSTT1. Carmustine 59-63 glutathione S-transferase theta 1 Homo sapiens 139-144 16874663-4 2006 It has been shown that also chemotherapeutic drugs such as BCNU [i.e. 1,3-bis(2-chloroethyl)-1-nitrosourea] are efficiently inactivated by GSTT1. Carmustine 70-106 glutathione S-transferase theta 1 Homo sapiens 139-144 17119257-8 2006 Variation in multiple benzene metabolizing genes may be associated with risk of benzene hematotoxicity, including CYP2E1, MPO, NQO1, and GSTT1. Benzene 80-87 glutathione S-transferase theta 1 Homo sapiens 137-142 16918316-3 2006 GSTT1-1 is highly conserved during evolution and plays a major role in phase-II biotransformation of a number of drugs and industrial chemicals, e.g. cytostatic drugs, hydrocarbons and halogenated hydrocarbons. Hydrocarbons 168-180 glutathione S-transferase theta 1 Homo sapiens 0-7 16938565-0 2006 Relation of glutathione S-transferase genotypes (GSTM1 and GSTT1) to laryngeal squamous cell carcinoma risk. Glutathione 12-23 glutathione S-transferase theta 1 Homo sapiens 59-64 16865249-0 2006 Potential application of GSTT1-null genotype in predicting toxicity associated to 5-fluouracil irinotecan and leucovorin regimen in advanced stage colorectal cancer patients. 5-fluouracil irinotecan 82-105 glutathione S-transferase theta 1 Homo sapiens 25-30 16865249-0 2006 Potential application of GSTT1-null genotype in predicting toxicity associated to 5-fluouracil irinotecan and leucovorin regimen in advanced stage colorectal cancer patients. Leucovorin 110-120 glutathione S-transferase theta 1 Homo sapiens 25-30 16865249-8 2006 Our data suggest that GSTT1-null is associated with a greater probability of developing toxicity to 5-Fu/CPT-11/Lv treatments, indicating a potential application of this genetic analysis in predicting adverse effects of this regimen. Fluorouracil 100-104 glutathione S-transferase theta 1 Homo sapiens 22-27 16865249-8 2006 Our data suggest that GSTT1-null is associated with a greater probability of developing toxicity to 5-Fu/CPT-11/Lv treatments, indicating a potential application of this genetic analysis in predicting adverse effects of this regimen. Irinotecan 105-111 glutathione S-transferase theta 1 Homo sapiens 22-27 16865249-8 2006 Our data suggest that GSTT1-null is associated with a greater probability of developing toxicity to 5-Fu/CPT-11/Lv treatments, indicating a potential application of this genetic analysis in predicting adverse effects of this regimen. Leucovorin 112-114 glutathione S-transferase theta 1 Homo sapiens 22-27 16918316-3 2006 GSTT1-1 is highly conserved during evolution and plays a major role in phase-II biotransformation of a number of drugs and industrial chemicals, e.g. cytostatic drugs, hydrocarbons and halogenated hydrocarbons. Hydrocarbons 197-209 glutathione S-transferase theta 1 Homo sapiens 0-7 16847185-13 2006 CONCLUSIONS: Our results suggest that the simultaneous presence of the GSTM1- and GSTT1-null genotypes is a susceptibility factor for DTC. dtc 134-137 glutathione S-transferase theta 1 Homo sapiens 82-87 16918316-5 2006 Several lines of evidence suggest that hGSTT1-1 and/or hGSTM1-1 play a role in the deactivation of reactive oxygen species that are likely to be involved in cellular processes of inflammation, ageing and degenerative diseases. Reactive Oxygen Species 99-122 glutathione S-transferase theta 1 Homo sapiens 39-47 16763966-0 2006 Nicotine metabolizing genes GSTT1 and CYP1A1 in sudden infant death syndrome. Nicotine 0-8 glutathione S-transferase theta 1 Homo sapiens 28-33 16898590-1 2006 In this study, we have investigated correlation between enzymatic activity of NAD(P)-dependent dehydrogenases of lymphocytes and polymorphic variants of glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) genes in the group of unrelated patients with acute pancreatitis in comparison with healthy Russians from Krasnoyarsk. NADP 78-84 glutathione S-transferase theta 1 Homo sapiens 198-203 16998606-3 2006 In our study we analysed the distribution of single and combined CYP1A1, GSTM1, GSTT1 and GSTP1 genotypes contributing to inter-individual differences in metabolism of xenobiotics and ROS in 125 Slovenian healthy individuals and in 140 patients with sporadic malignant melanoma. Reactive Oxygen Species 184-187 glutathione S-transferase theta 1 Homo sapiens 80-85 16424821-5 2006 The possible influence of genetic polymorphisms of xenobiotic-metabolizing enzymes involved in styrene biotransformation (EPHX1, GSTT1, GSTM1, GSTP1) and NAT2 on the cytogenetic endpoints was investigated. Styrene 95-102 glutathione S-transferase theta 1 Homo sapiens 129-134 16753877-4 2006 In this study, the expression of wild and mutant type GSTT-1 gene of those stable transformants and bone marrow cells from MDS patients by RT-PCR was observed in the presence or absence of rapamycin. Sirolimus 189-198 glutathione S-transferase theta 1 Homo sapiens 54-60 16624155-5 2006 The risk of failure to achieve CR in patients with GSTT1 null/GSTM1 null is 8.736 times higher than that in patients with GSTT1 and GSTM1 genes double-present (odds ratio OR was 8.736, 95% CI was 1.146 - 66.574). Chromium 31-33 glutathione S-transferase theta 1 Homo sapiens 51-56 16424821-12 2006 Our results suggest that occupational exposure to styrene has genotoxic effects that are potentiated by the GSTT1 gene deletion. Styrene 50-57 glutathione S-transferase theta 1 Homo sapiens 108-113 16125881-4 2005 The subjects" genetic polymorphisms in the genes that encode the styrene-metabolizing enzymes CYP2E1, CYP2B6, EPHX1, GSTM1, GSTT1 and GSTP1 were determined. Styrene 65-72 glutathione S-transferase theta 1 Homo sapiens 124-129