Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 161
Filtrar
2.
Nat Commun ; 12(1): 326, 2021 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-33436607

RESUMO

Adipose tissue-resident T cells have been recognized as a critical regulator of thermogenesis and energy expenditure, yet the underlying mechanisms remain unclear. Here, we show that high-fat diet (HFD) feeding greatly suppresses the expression of disulfide-bond A oxidoreductase-like protein (DsbA-L), a mitochondria-localized chaperone protein, in adipose-resident T cells, which correlates with reduced T cell mitochondrial function. T cell-specific knockout of DsbA-L enhances diet-induced thermogenesis in brown adipose tissue (BAT) and protects mice from HFD-induced obesity, hepatosteatosis, and insulin resistance. Mechanistically, DsbA-L deficiency in T cells reduces IFN-γ production and activates protein kinase A by reducing phosphodiesterase-4D expression, leading to increased BAT thermogenesis. Taken together, our study uncovers a mechanism by which T cells communicate with brown adipocytes to regulate BAT thermogenesis and whole-body energy homeostasis. Our findings highlight a therapeutic potential of targeting T cells for the treatment of over nutrition-induced obesity and its associated metabolic diseases.


Assuntos
Dieta Hiperlipídica , Glutationa Transferase/deficiência , Interferon gama/biossíntese , Linfócitos T/metabolismo , Termogênese , Adipócitos Marrons/efeitos dos fármacos , Adipócitos Marrons/metabolismo , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/metabolismo , Tecido Adiposo Branco/efeitos dos fármacos , Tecido Adiposo Branco/metabolismo , Animais , Regulação para Baixo/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Comportamento Alimentar , Glutationa Transferase/metabolismo , Resistência à Insulina , Interferon gama/administração & dosagem , Interferon gama/farmacologia , Masculino , Camundongos Knockout , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Obesidade/genética , Obesidade/patologia , Linfócitos T/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/metabolismo , Termogênese/efeitos dos fármacos , Termogênese/genética , Proteína Desacopladora 1/metabolismo
3.
Biochem Biophys Res Commun ; 528(3): 538-544, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32507596

RESUMO

Glutathione S-transferases (GSTs) play important roles in metabolism and detoxification of plant cells. However, their functions during development are less well understood. Arabidopsis AtGSTU7 (AT2G29420) encodes a Tau class GST. Here we provide the AtGSTU7 was abundantly expressed in seeds and in roots at an early stage of germination. AtGSTU7 expression was repressed by exogenous ABA and promoted by osmotic stress. A null mutant of AtGSTU7 (atgstu7) accumulated higher contents of reduced GSH and decreased amounts of endogenous H2O2 in seedlings. The atgstu7 plants showed decreased osmotic tolerance during seed germination, which was influenced by GSH and ABI3 gene expression. The results suggested that AtGSTU7 involvement in seed germination is mediated by GSH-ROS homeostasis and ABA signaling.


Assuntos
Proteínas de Arabidopsis/genética , Arabidopsis/genética , Arabidopsis/metabolismo , Glutationa Transferase/genética , Ácido Abscísico/metabolismo , Ácido Abscísico/farmacologia , Arabidopsis/crescimento & desenvolvimento , Proteínas de Arabidopsis/antagonistas & inibidores , Proteínas de Arabidopsis/metabolismo , Regulação da Expressão Gênica de Plantas , Técnicas de Inativação de Genes , Genes de Plantas , Germinação/genética , Germinação/fisiologia , Glutationa/metabolismo , Glutationa Transferase/deficiência , Glutationa Transferase/metabolismo , Pressão Osmótica , Reguladores de Crescimento de Plantas/metabolismo , Reguladores de Crescimento de Plantas/farmacologia , Plantas Geneticamente Modificadas , Espécies Reativas de Oxigênio/metabolismo , Sementes/genética , Sementes/crescimento & desenvolvimento , Sementes/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
Chem Res Toxicol ; 33(7): 1698-1708, 2020 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-32237725

RESUMO

Smoking-induced lung cancer is a major cause of cancer mortality in the US and worldwide. While 11-24% of smokers will develop lung cancer, risk varies among individuals and ethnic/racial groups. Specifically, African American and Native Hawaiian cigarette smokers are more likely to get lung cancer as compared to Caucasians, Japanese Americans, and Latinos. It is important to identify smokers who are at the greatest risk of developing lung cancer as they should be candidates for smoking cessation and chemopreventive intervention programs. Among 60+ tobacco smoke carcinogens, 1,3-butadiene (BD) is one of the most potent and abundant (20-75 µg per cigarette in mainstream smoke and 205-361 µg per cigarette in side stream smoke). 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 7-(1-hydroxy-3-buten-2-yl)guanine (EB-GII) adducts. In the present study, we employed EBV-transformed human lymphoblastoid cell lines (HapMap cells) with known GSTT1 genotypes to examine the influence of the GSTT1 gene on interindividual variability in butadiene metabolism, DNA adduct formation/repair, and biological outcomes (apoptosis). 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 as GSTT1+ cells. Our results suggest that GSTT1 can influence an individual's susceptibility to butadiene-derived epoxides.


Assuntos
Butadienos/metabolismo , Adutos de DNA/biossíntese , Compostos de Epóxi/metabolismo , Glutationa Transferase/metabolismo , Guanina/biossíntese , Haplótipos , Apoptose , Butadienos/química , Linhagem Celular , Reparo do DNA , Compostos de Epóxi/química , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Guanina/análogos & derivados , Humanos , Estrutura Molecular
5.
Clin Sci (Lond) ; 134(7): 677-694, 2020 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-32167139

RESUMO

Excessive mitochondrial fission has been identified as the central pathogenesis of diabetic kidney disease (DKD), but the precise mechanisms remain unclear. Disulfide-bond A oxidoreductase-like protein (DsbA-L) is highly expressed in mitochondria in tubular cells of the kidney, but its pathophysiological role in DKD is unknown. Our bioinformatics analysis showed that tubular DsbA-L mRNA levels were positively associated with eGFR but negatively associated with Scr and 24h-proteinuria in CKD patients. Furthermore, the genes that were coexpressed with DsbA-L were mainly enriched in mitochondria and were involved in oxidative phosphorylation. In vivo, knockout of DsbA-L exacerbated diabetic mice tubular cell mitochondrial fragmentation, oxidative stress and renal damage. In vitro, we found that DsbA-L was localized in the mitochondria of HK-2 cells. High glucose (HG, 30 mM) treatment decreased DsbA-L expression followed by increased mitochondrial ROS (mtROS) generation and mitochondrial fragmentation. In addition, DsbA-L knockdown exacerbated these abnormalities, but this effect was reversed by overexpression of DsbA-L. Mechanistically, under HG conditions, knockdown DsbA-L expression accentuated JNK phosphorylation in HK-2 cells. Furthermore, administration of a JNK inhibitor (SP600125) or the mtROS scavenger MitoQ significantly attenuated JNK activation and subsequent mitochondrial fragmentation in DsbA-L-knockdown HK-2 cells. Additionally, the down-regulation of DsbA-L also amplified the gene and protein expression of mitochondrial fission factor (MFF) via the JNK pathway, enhancing its ability to recruit DRP1 to mitochondria. Taken together, these results link DsbA-L to alterations in mitochondrial dynamics during tubular injury in the pathogenesis of DKD and unveil a novel mechanism by which DsbA-L modifies mtROS/JNK/MFF-related mitochondrial fission.


Assuntos
Diabetes Mellitus Experimental/enzimologia , Nefropatias Diabéticas/enzimologia , Glutationa Transferase/deficiência , Túbulos Renais/enzimologia , Mitocôndrias/enzimologia , Dinâmica Mitocondrial , Animais , Glicemia/metabolismo , Linhagem Celular , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/etiologia , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Glutationa Transferase/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Túbulos Renais/ultraestrutura , Proteínas de Membrana/metabolismo , Camundongos Knockout , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/metabolismo , Estresse Oxidativo , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais
6.
Chin Med Sci J ; 34(3): 205-210, 2019 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-31648673

RESUMO

Objective To clarify the possible association of GSTT1 homozygous deletion with the susceptibility to pancreatic cancer. Methods We searched PubMed database, Chinese Journal Full Text Database (CNKI), and EMBASE to find the eligible studies published up to April 18, 2018 for evaluating the relationship between GSTT1 homozygous deletion and pancreatic cancer. The frequency of null genotype for GSTT1 between the pancreatic cancer group and the healthy control group was compared with Chi-square test, and odds ratios (ORs) value and 95% confidence interval (95% CI) were calculated. Results A total of 9 studies met the inclusion criteria, and 5952 cases consisting of 2387 pancreatic cancer patients and 3565 healthy controls were included in the meta analysis. Compared with the control group, frequency of null genotype for GSTT1 in the pancreatic cancer group was higher (33.4% vs. 38.7%, OR = 1.26, 95%CI = 1.01-1.58, P = 0.04). Conclusion GSTT1 homozygous deletion individuals may have higher susceptibility to pancreatic cancer.


Assuntos
Deleção de Genes , Predisposição Genética para Doença , Glutationa Transferase/deficiência , Homozigoto , Neoplasias Pancreáticas/genética , Humanos , Neoplasias Pancreáticas/enzimologia
7.
Nat Commun ; 10(1): 4150, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515474

RESUMO

Cisplatin is one of the most widely used chemotherapeutic drugs for the treatment of cancer. Unfortunately, one of its major side effects is permanent hearing loss. Here, we show that glutathione transferase α4 (GSTA4), a member of the Phase II detoxifying enzyme superfamily, mediates reduction of cisplatin ototoxicity by removing 4-hydroxynonenal (4-HNE) in the inner ears of female mice. Under cisplatin treatment, loss of Gsta4 results in more profound hearing loss in female mice compared to male mice. Cisplatin stimulates GSTA4 activity in the inner ear of female wild-type, but not male wild-type mice. In female Gsta4-/- mice, cisplatin treatment results in increased levels of 4-HNE in cochlear neurons compared to male Gsta4-/- mice. In CBA/CaJ mice, ovariectomy decreases mRNA expression of Gsta4, and the levels of GSTA4 protein in the inner ears. Thus, our findings suggest that GSTA4-dependent detoxification may play a role in estrogen-mediated neuroprotection.


Assuntos
Cisplatino/efeitos adversos , Glutationa Transferase/metabolismo , Ototoxicidade/enzimologia , Animais , Limiar Auditivo/efeitos dos fármacos , Capilares/patologia , Cóclea/enzimologia , Cóclea/patologia , Cóclea/fisiopatologia , Cruzamentos Genéticos , Dano ao DNA/genética , Potenciais Evocados Auditivos do Tronco Encefálico/efeitos dos fármacos , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Glutationa Transferase/deficiência , Perda Auditiva/complicações , Perda Auditiva/enzimologia , Perda Auditiva/fisiopatologia , Masculino , Camundongos Endogâmicos CBA , Ototoxicidade/complicações , Ototoxicidade/patologia , Ototoxicidade/fisiopatologia , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Gânglio Espiral da Cóclea/efeitos dos fármacos , Gânglio Espiral da Cóclea/patologia
8.
Pest Manag Sci ; 75(6): 1527-1538, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30610767

RESUMO

BACKGROUND: The oriental fruit fly Bactrocera dorsalis (Hendel), a widespread agricultural pest, has evolved resistance to many insecticides, including organophosphorus compounds. Glutathione S-transferases (GSTs) are involved in xenobiotic detoxification and insecticide resistance in many insects. However, the role of delta class GSTs in detoxifying malathion in B. dorsalis is unknown. Here, we evaluated the roles of two delta class GSTs in malathion detoxification in this species. RESULTS: Two delta class GSTs genes, BdGSTd1 and BdGSTd10, were characterized in B. dorsalis. They were highly expressed in 5-day-old adults, as well as in midgut and Malpighian tubules. Upon malathion exposure, the two genes were upregulated by 2.63- and 2.85-fold, respectively. Injection of double-stranded RNA targeting BdGSTd1 or BdGSTd10 significantly reduced their mRNA levels in adults and also significantly increased adult susceptibility to malathion. The expression of these two GSTs in Escherichia coli helped the host to endure malathion stress at a concentration of 10 µg mL-1 according to a Cell Counting Kit-8 assay. High-performance liquid chromatography analyses indicated that malathion could be significantly depleted by the two delta GSTs. The role of BdGSTd10 in malathion sequestration was also discussed. CONCLUSION: BdGSTd1 and BdGSTd10 play important roles in the detoxification of malathion in B. dorsalis. © 2019 Society of Chemical Industry.


Assuntos
Glutationa Transferase/metabolismo , Inativação Metabólica , Malation/metabolismo , Tephritidae/metabolismo , Sequência de Aminoácidos , Animais , Regulação Enzimológica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glutationa Transferase/química , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Cinética , Malation/toxicidade , Filogenia , Tephritidae/enzimologia
9.
Nat Commun ; 9(1): 4815, 2018 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-30446643

RESUMO

Common disorders, including diabetes and Parkinson's disease, are caused by a combination of environmental factors and genetic susceptibility. However, defining the mechanisms underlying gene-environment interactions has been challenging due to the lack of a suitable experimental platform. Using pancreatic ß-like cells derived from human pluripotent stem cells (hPSCs), we discovered that a commonly used pesticide, propargite, induces pancreatic ß-cell death, a pathological hallmark of diabetes. Screening a panel of diverse hPSC-derived cell types we extended this observation to a similar susceptibility in midbrain dopamine neurons, a cell type affected in Parkinson's disease. We assessed gene-environment interactions using isogenic hPSC lines for genetic variants associated with diabetes and Parkinson's disease. We found GSTT1-/- pancreatic ß-like cells and dopamine neurons were both hypersensitive to propargite-induced cell death. Our study identifies an environmental chemical that contributes to human ß-cell and dopamine neuron loss and validates a novel hPSC-based platform for determining gene-environment interactions.


Assuntos
Cicloexanos/toxicidade , Diabetes Mellitus/induzido quimicamente , Neurônios Dopaminérgicos/efeitos dos fármacos , Interação Gene-Ambiente , Células Secretoras de Insulina/efeitos dos fármacos , Praguicidas/toxicidade , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Diferenciação Celular , Diabetes Mellitus/genética , Diabetes Mellitus/patologia , Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/enzimologia , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/enzimologia , Mesencéfalo/citologia , Mesencéfalo/efeitos dos fármacos , Mesencéfalo/enzimologia , Camundongos , Modelos Biológicos , Doença de Parkinson/etiologia , Doença de Parkinson/genética , Doença de Parkinson/patologia , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/enzimologia
10.
Alcohol Clin Exp Res ; 42(7): 1192-1205, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29708596

RESUMO

BACKGROUND: Glutathione S-transferase A4-4 (GSTA4) is a key enzyme for removal of toxic lipid peroxidation products such as 4-hydroxynonenal (4-HNE). In this study, we examined the potential role of GSTA4 on protein carbonylation and progression of alcoholic liver disease by examining the development of liver injury in male wild-type (WT) SV/J mice and SV/J mice lacking functional GSTA4 (GSTA4-/- mice). METHODS: Adult male WT and GSTA4-/- mice were fed chow (N = 10 to 12) or high-fat Lieber-DeCarli liquid diets containing up to 28% calories as ethanol (EtOH) (N = 18 to 20) for 116 days. At the end of the study, half of the EtOH-fed mice were acutely challenged with an EtOH binge (3 g/kg given intragastrically) 12 hours before sacrifice. Carbonylation of liver proteins was assessed by immunohistochemical staining for 4-HNE adduction and by comprehensive liquid chromatography-tandem mass spectrometry (LC-MS/MS) of purified carbonylated proteins. RESULTS: Chronic EtOH intake significantly increased hepatic 4-HNE adduction and protein carbonylation, including carbonylation of ribosomal proteins. EtOH intake also resulted in steatosis and increased serum alanine aminotransferase. Hepatic infiltration with B cells, T cells, and neutrophils and mRNA expression of pro-inflammatory cytokines tumor necrosis factor (TNF)α and interferon (IFN)γ was modest in WT mice. However, an EtOH binge increased hepatic necrosis, hepatic cell proliferation, and expression of TNFα mRNA (p < 0.05). EtOH treatment of GSTA4-/- mice increased B-cell infiltration and increased mRNA expression of TNFα and IFNγ and of matrix remodeling markers MMP9, MMP13, and Col1A1 (p < 0.05). GSTA4-/- mice exhibited panlobular rather than periportal distribution of 4-HNE-adducted proteins and increased overall 4-HNE staining after EtOH binge. Comprehensive LC-MS of carbonylated proteins identified 1,022 proteins of which 189 were unique to the GSTA4-/- group. CONCLUSIONS: These data suggest long-term adaptation to EtOH in WT mice does not occur in GSTA4-/- mice. Products of lipid peroxidation appear to play a role in inflammatory responses due to EtOH. And EtOH effects on B-cell infiltration and autoimmune responses may be secondary to formation of carbonyl adducts.


Assuntos
Etanol/toxicidade , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Hepatopatias Alcoólicas/genética , Hepatopatias Alcoólicas/metabolismo , Carbonilação Proteica/fisiologia , Animais , Etanol/administração & dosagem , Glutationa Transferase/química , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Hepatopatias Alcoólicas/patologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Knockout , Carbonilação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
11.
Proc Natl Acad Sci U S A ; 114(46): 12196-12201, 2017 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-29087318

RESUMO

Chronic inflammation in adipose tissue plays a key role in obesity-induced insulin resistance. However, the mechanisms underlying obesity-induced inflammation remain elusive. Here we show that obesity promotes mtDNA release into the cytosol, where it triggers inflammatory responses by activating the DNA-sensing cGAS-cGAMP-STING pathway. Fat-specific knockout of disulfide-bond A oxidoreductase-like protein (DsbA-L), a chaperone-like protein originally identified in the mitochondrial matrix, impaired mitochondrial function and promoted mtDNA release, leading to activation of the cGAS-cGAMP-STING pathway and inflammatory responses. Conversely, fat-specific overexpression of DsbA-L protected mice against high-fat diet-induced activation of the cGAS-cGAMP-STING pathway and inflammation. Taken together, we identify DsbA-L as a key molecule that maintains mitochondrial integrity. DsbA-L deficiency promotes inflammation and insulin resistance by activating the cGAS-cGAMP-STING pathway. Our study also reveals that, in addition to its well-characterized roles in innate immune surveillance, the cGAS-cGAMP-STING pathway plays an important role in mediating obesity-induced metabolic dysfunction.


Assuntos
DNA Mitocondrial/metabolismo , Glutationa Transferase/genética , Resistência à Insulina , Proteínas de Membrana/genética , Nucleotidiltransferases/genética , Obesidade/genética , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/patologia , Animais , Dieta Hiperlipídica/efeitos adversos , Regulação da Expressão Gênica , Glutationa Transferase/deficiência , Humanos , Inflamação , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Nucleotidiltransferases/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Obesidade/patologia , Cultura Primária de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transdução de Sinais
12.
BMJ Case Rep ; 20172017 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-28069791

RESUMO

A 33-week gestation boy with Mediterranean glucose-6-phosphate dehydrogenase (G6PD) and a glutathione S-transferase Mu 1 null mutations (GSTM1*0/*0) developed prolonged indirect hyperbilirubinemia (PIH). He had no laboratory evidence of haemolysis or infection, and no exposure to oxidising agents. He has two full-term older brothers who have no history of neonatal hyperbilirubinemia. One brother, who was exclusively breast fed, has only Mediterranean G6PD and the other has only GSTM1*0/*0. The three boys have no mutation in the uridine diphosphate glucuronosyltransferase 1A1 gene. This suggests that a combination of all or any two of prematurity, G6PD deficiency and GSTM1*0/*0 is a possible risk factor for PIH. However, this remains to be confirmed.


Assuntos
Deficiência de Glucosefosfato Desidrogenase/genética , Glutationa Transferase/genética , Hiperbilirrubinemia Neonatal/genética , Mutação/genética , Assistência ao Convalescente , Diagnóstico Diferencial , Glutationa Transferase/deficiência , Humanos , Hiperbilirrubinemia Neonatal/terapia , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Fototerapia
13.
Am J Perinatol ; 34(3): 305-314, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27464020

RESUMO

Classically, genetically decreased bilirubin conjugation and/or hemolysis account for the mechanisms contributing to neonatal hyperbilirubinemia associated with glucose-6-phosphate dehydrogenase (G6PD) deficiency. However, these mechanisms are not involved in most cases of this hyperbilirubinemia. Additional plausible mechanisms for G6PD deficiency-associated hyperbilirubinemia need to be considered. Glutathione S-transferases (GST) activity depends on a steady quantity of reduced form of glutathione (GSH). If GSH is oxidized, it is reduced back by glutathione reductase, which requires the reduced form of nicotinamide adenine dinucleotide phosphate (NADPH). The main source of NADPH is the pentose phosphate pathway, in which G6PD is the first enzyme. Rat kidney GSH, rat liver GST, and human red blood cell GST levels have been found to positively correlate with G6PD levels in their respective tissues. As G6PD is expressed in hepatocytes, it is expected that GST levels would be significantly decreased in hepatocytes of G6PD-deficient neonates. As hepatic GST binds bilirubin and prevents their reflux into circulation, hypothesis that decreased GST levels in hepatocytes is an additional mechanism contributing to G6PD deficiency-associated hyperbilirubinemia seems plausible. Evidence for and against this hypothesis are discussed in this article hoping to stimulate further research on the role of GST in G6PD deficiency-associated hyperbilirubinemia.


Assuntos
Deficiência de Glucosefosfato Desidrogenase/metabolismo , Glutationa Transferase/metabolismo , Hepatócitos/enzimologia , Hiperbilirrubinemia Neonatal/metabolismo , Animais , Bilirrubina/análogos & derivados , Bilirrubina/sangue , Eritrócitos/enzimologia , Doença de Gilbert/metabolismo , Deficiência de Glucosefosfato Desidrogenase/complicações , Glutationa Transferase/deficiência , Glutationa Transferase/genética , Humanos , Hiperbilirrubinemia Neonatal/complicações , Recém-Nascido
14.
Am J Pathol ; 187(2): 418-430, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27998724

RESUMO

We established a mouse model of developmental nonalcoholic steatohepatitis (NASH) by feeding a high polyunsaturated fat liquid diet to female glutathione-S-transferase 4-4 (Gsta4-/-)/peroxisome proliferator activated receptor α (Ppara-/-) double knockout 129/SvJ mice for 12 weeks from weaning. We used it to probe the importance of lipid peroxidation in progression of NASH beyond simple steatosis. Feeding Gsta4-/-/Ppara-/- double-knockout (dKO) mice liquid diets containing corn oil resulted in a percentage fat-dependent increase in steatosis and necroinflammatory injury (P < 0.05). Increasing fat to 70% from 35% resulted in increases in formation of 4-hydroxynonenal protein adducts accompanied by evidence of stellate cell activation, matrix remodeling, and fibrosis (P < 0.05). Comparison of dKO mice with wild-type (Wt) and single knockout mice revealed additive effects of Gsta4-/- and Ppara-/- silencing on steatosis, 4-hydroxynonenal adduct formation, oxidative stress, serum alanine amino transferase, expression of tumor necrosis factor alpha, Il6, interferon mRNA, and liver pathology (P < 0.05). Induction of Cyp2e1 protein by high-fat diet was suppressed in Gsta4-/- and dKO groups (P < 0.05). The dKO mice had similar levels of markers of stellate cell activation and matrix remodeling as Ppara-/- single KO mice. These data suggest that lipid peroxidation products play a role in progression of liver injury to steatohepatitis in NASH produced by high-fat feeding during development but appear less important in development of fibrosis.


Assuntos
Glutationa Transferase/deficiência , Peroxidação de Lipídeos/fisiologia , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/patologia , Animais , Western Blotting , Dieta Hiperlipídica , Modelos Animais de Doenças , Feminino , Fígado/patologia , Camundongos , Camundongos Knockout , PPAR alfa/deficiência , Reação em Cadeia da Polimerase , Transcriptoma
15.
J Med Genet ; 54(4): 241-247, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-27876694

RESUMO

BACKGROUND: A high level of succinylacetone (SA) in blood is a sensitive, specific newborn screening marker for hepatorenal tyrosinemia type 1 (HT1, MIM 276700) caused by deficiency of fumarylacetoacetate hydrolase (FAH). Newborns with HT1 are usually clinically asymptomatic but show liver dysfunction with coagulation abnormalities (prolonged prothrombin time and/or high international normalised ratio). Early treatment with nitisinone (NTBC) plus dietary restriction of tyrosine and phenylalanine prevents the complications of severe liver disease and neurological crises. METHODS AND RESULTS: Six newborns referred for hypersuccinylacetonaemia but who had normal coagulation testing on initial evaluation had sequence variants in the GSTZ1 gene, encoding maleylacetoacetate isomerase (MAAI), the enzyme preceding FAH in tyrosine degradation. Initial plasma SA levels ranged from 233 to 1282 nmol/L, greater than normal (<24 nmol/L) but less than the initial values of patients with HT1 (16 944-74 377 nmol/L, n=15). Four individuals were homozygous for c.449C>T (p.Ala150Val). One was compound heterozygous for c.259C>T (p.Arg87Ter) and an intronic sequence variant. In one, a single heterozygous GSTZ1 sequence variant was identified, c.295G>A (p.Val99Met). Bacterial expression of p.Ala150Val and p.Val99Met revealed low MAAI activity. The six individuals with mild hypersuccinylacetonaemia (MHSA) were not treated with diet or nitisinone. Their clinical course has been normal for up to 13 years. CONCLUSIONS: MHSA can be caused by sequence variants in GSTZ1. Such individuals have thus far remained asymptomatic despite receiving no specific treatment.


Assuntos
Glutationa Transferase/genética , Hidrolases/genética , Fígado/enzimologia , Tirosinemias/genética , Adolescente , Criança , Pré-Escolar , Feminino , Variação Genética , Glutationa Transferase/deficiência , Heptanoatos/sangue , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Hidrolases/sangue , Lactente , Recém-Nascido , Fígado/patologia , Masculino , Tirosina/sangue , Tirosinemias/sangue , Tirosinemias/patologia
16.
Sci Rep ; 6: 38254, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27905550

RESUMO

Glutathione-S-transferases mu 2 (GSTM2), a kind of important Phase II antioxidant enzyme of eukaryotes, is degraded by nonsense mediated mRNA decay due to a C27T substitution in the fifth exon of pigs. As a reproductive performance-related gene, GSTM2 is involved in embryo implantation, whereas, functional deficiency of GSTM2 induces pre- or post-natal death in piglets potentially. To have some insight into the role of GSTM2 in embryo development, high throughput RNA sequencing is performed using the swine testis cells (ST) with the deletion of GSTM2. Some embryo development-related genes are observed from a total of 242 differentially expressed genes, including STAT1, SRC, IL-8, DUSP family, CCL family and integrin family. GSTM2 affects expression of SRC, OPN, and SLCs. GSTM2 suppresses phosphorylation of STAT1 by binding to STAT1. In addition, as an important transcription factor, STAT1 regulates expression of uterus receptive-related genes including CCLs, IRF9, IFITs, MXs, and OAS. The present study provides evidence to molecular mechanism of GSTM2 modulating embryo development.


Assuntos
Embrião de Mamíferos/enzimologia , Desenvolvimento Embrionário , Perfilação da Expressão Gênica , Glutationa Transferase/deficiência , Sequenciamento de Nucleotídeos em Larga Escala , Testículo/enzimologia , Transcriptoma , Animais , Técnicas de Silenciamento de Genes , Glutationa Transferase/metabolismo , Masculino , Suínos , Testículo/embriologia
17.
Genet Mol Res ; 15(3)2016 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-27706600

RESUMO

The aim of this study was to determine the prevalence of polymorphisms in the glutathione S-transferase genes GSTM1 and GSTT1 in patients with lens opacity (cataract). Peripheral blood samples were obtained from male and female patients (N = 23) with cataract. The GSTM1 and GSTT1 polymorphic regions were amplified by polymerase chain reaction, and the amplification products were electrophoresed on a 2% agarose gel. The obtained bands were by staining with ethidium bromide. The results were compared by a chi-square test using the BioEstat software (v.5.0). The frequencies of the GSTM1- and GSTT1-null genotypes were higher than those of the GSTM1- and GSTT1-present genotypes. The frequency of GSTT1-null genotypes was approximately 1.7 times higher than that of GSTM1, which was a statistically significant difference (P = 0.0019). Although a consensus remains to be reached on the correlation between genetic polymorphisms in GSTs and cataract susceptibility, the observations from most scientific studies are similar to those reported in this study. Thus, we conclude that the absence of these genes, particularly GSTT1, is correlated with the development of lens opacity.


Assuntos
Catarata/diagnóstico , Catarata/genética , Predisposição Genética para Doença , Glutationa Transferase/genética , Polimorfismo Genético , Adulto , Catarata/patologia , Feminino , Expressão Gênica , Frequência do Gene , Glutationa Transferase/deficiência , Humanos , Cristalino/metabolismo , Cristalino/patologia , Masculino , Pessoa de Meia-Idade
18.
Genet Mol Res ; 15(1)2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26909940

RESUMO

The enzyme glutathione S-transferase Mu 1 (GSTM1) is encoded by the GSTM1 gene. Polymorphisms in GSTM1 affect the detoxifying function of the enzyme variants. This forms the basis of the debate about the impact of the GSTM1 null/present genotype on colorectal carcinoma risk. We tested the potential influence of GSTM1 polymorphisms on the development of colorectal cancer. DNA extracted from 83 samples taken from patients that were previously diagnosed as having colorectal carcinoma and from 35 control subjects who did not have colorectal carcinoma were amplified. GSTM1 genotypes were determined by DNA sequencing. The current study revealed that the majority (69/83, 83%) of colorectal cancer cases harbored the null genotype (GSTM1*0/*0), and the remaining 14 (17%) cases harbored either the GSTM1wt/wt or the GSTM1wt/*0 genotype. In contrast, among the control cases, 23 (65%) had the null genotype (GSTM1*0/*0) and 12 (35%) had either the GSTM1wt/wt or the GSTM1wt/*0 genotype. The current report emphasizes the impact of the GSTM1 null genotype on the increased risk of colorectal carcinoma in Saudi Arabia.


Assuntos
Carcinoma/genética , Neoplasias Colorretais/genética , Predisposição Genética para Doença , Glutationa Transferase/genética , Polimorfismo Genético , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma/diagnóstico , Carcinoma/patologia , Estudos de Casos e Controles , Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/patologia , Feminino , Expressão Gênica , Glutationa Transferase/deficiência , Humanos , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Arábia Saudita , Análise de Sequência de DNA
19.
J Obstet Gynaecol Res ; 42(4): 434-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26785830

RESUMO

AIM: Few studies have investigated the role of the GSTM1 and GSTT1 genes in uterine leiomyoma. Therefore, in the current study the distribution of these genotypes in Iranian women and susceptibility to uterine leiomyoma was investigated. METHODS: Blood samples of 50 patients with uterine leiomyoma and 50 healthy individual controls were collected in this cross-sectional study. Genomic DNA was extracted, and subsequently GSTM1 and GSTT1 null genotypes were detected by the Gap-polymerase chain reaction method. RESULTS: A total of 42% of patients appeared to lack GSTM1 enzyme activity due to the presence of an extended deletion (GSTM1 0/0 genotype), compared with 18% in a control group (odds ratio [OR], 3.56; 95% confidence interval [CI], 1.35-9.37; P < 0.010). In addition, the prevalence of the GSTT1 null genotype in patients was higher than that in the control group (42% to 14%, P < 0.009). Also, it was shown that individuals with both null genotypes (-/-) had a 19.23-fold higher risk of developing the disease in comparison to people who showed both present genotypes (+/+). (P = 0.007; 95%CI, 2.20-167.41). Besides, it was observed that at least one null genotype increases the risk of myoma to 2.6 compared to the both present genotype (P-value < 0.03, 95%CI, 1.05-6.82). CONCLUSION: To our knowledge, this is first significant correlation between risk of uterine leiomyoma and null GSTM1 and GSTT1 genotypes among Iranian patients. Our data support the involvement of GSTM1 and GSTT1 in uterine leiomyoma liability, and especially its role as a genetic factor in the occurrence of this disease.


Assuntos
Deleção de Genes , Predisposição Genética para Doença , Glutationa Transferase/genética , Leiomioma/genética , Neoplasias Uterinas/genética , Adolescente , Adulto , Estudos Transversais , Feminino , Genótipo , Glutationa Transferase/deficiência , Humanos , Irã (Geográfico) , Pessoa de Meia-Idade , Razão de Chances , Polimorfismo Genético
20.
Hematology ; 21(1): 46-53, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26288192

RESUMO

OBJECTIVES: Estimating the prevalence of glutathione S-transferase gene polymorphism (GSTM1) null genotype among patients with beta thalassemia major (ß-TM) in relation to myocardial status assessed by tissue Doppler and cardiac siderosis assessed by cardiac magnetic resonance imaging (MRI) T2*. METHODS: Hundred patients with ß-TM and 100 healthy controls were enrolled. Complete blood count (CBC), mean serum ferritin and GSTM1 genotyping, echocardiography, tissue Doppler, and cardiac MRI T2* were done. RESULTS: Serum ferritin ranged from 1200 to 8000 ng/ml, and mean T2* value was 27.10 ± 11.20 ms. Of patients, 68 (68%) had no cardiac siderosis, while 24 (24%) with mild to moderate, and 8 (8%) with sever cardiac siderosis. T2* values were not correlated with serum ferritin (r = -0.09, P = 0.50). GSTM1 null genotype was prevalent in 46% of patients and 40% of controls (P = 0.69). Patients with null genotype had significantly shorter T2* (P = 0.001), higher left ventricular end-diastolic diameter (P = 0.002), and shorter ejection time (P = 0.005) with no significant relation to serum ferritin (P = 0.122). GSTM1 null genotype was the only predictor for cardiac iron overload (P = 0.002). DISCUSSION: Serum ferritin concentrations have been shown to correlate poorly with all stages of cardiac dysfunction. Low cardiac MRI T2* values occur in patients with ß-TM despite good chelation therapy, suggesting a possible role of genetic factors in cardiac siderosis. CONCLUSION: GSTM1 null genotype is significantly associated with cardiac iron overload independent of serum ferritin in Egyptian patients with ß-TM.


Assuntos
Glutationa Transferase/genética , Sobrecarga de Ferro/genética , Ferro/metabolismo , Polimorfismo Genético , Siderose/genética , Talassemia beta/terapia , Adolescente , Estudos de Casos e Controles , Criança , Egito , Feminino , Ferritinas/sangue , Ferritinas/genética , Expressão Gênica , Genótipo , Glutationa Transferase/deficiência , Humanos , Sobrecarga de Ferro/etiologia , Sobrecarga de Ferro/metabolismo , Sobrecarga de Ferro/patologia , Masculino , Miocárdio/metabolismo , Miocárdio/patologia , Índice de Gravidade de Doença , Siderose/etiologia , Siderose/metabolismo , Siderose/patologia , Reação Transfusional , Talassemia beta/genética , Talassemia beta/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...