1
|
Wang M, Shen Y, Gao Y, Chen H, Duan F, Li S, Wang G. NQO1 polymorphism and susceptibility to ischemic stroke in a Chinese population. BMC Med Genomics 2024; 17:219. [PMID: 39174970 PMCID: PMC11342592 DOI: 10.1186/s12920-024-01992-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Ischemic stroke (IS) is a major cause of death and disability worldwide. Genetic factors are important risk factors for the development of IS. The quinone oxidoreductase 1 gene (NQO1) has antioxidant, anti-inflammatory, and cytoprotective properties. Thus, in this study, we investigated the relationship between NQO1 gene polymorphism and the risk of IS. METHODS Peripheral blood was collected from 143 patients with IS and 124 the control groups in Yunnan, China, and NQO1 rs2917673, rs689455, and rs1800566 were genotyped. Logistic regression was used to analyze the relationship between the three NQO1 loci and IS susceptibility. The difference in the expression levels of NQO1 between the control groups and IS groups was verified using public databases and enzyme-linked immunosorbent assay. RESULTS The rs2917673 locus increased the risk of IS by 2.375 times in TT genotype carriers under the co-dominance model compared with CC carriers and was statistically associated with the risk of IS (OR = 2.375, 95% CI = 1.017-5.546, P = 0.046). In the recessive model, TT genotype carriers increased IS risk by 2.407 times compared with CC/CT carriers and were statistically associated with the risk of IS (OR = 2.407, 95% CI = 1.073-5.396, P = 0.033). CONCLUSIONS NQO1 rs2917673 polymorphism is significantly associated with IS. Mutant TT carriers are risk factors for IS.
Collapse
Affiliation(s)
- Min Wang
- School of Clinical Medicine, Dali University, Dali, Yunnan, 671000, PR China
| | - Ying Shen
- The First Hospital of Liangshan, Xichang, Sichuan, 615000, PR China
| | - Yuan Gao
- School of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Huaqiu Chen
- Xichang People's Hospital, Xichang, Sichuan, 615000, PR China
| | - Fuhui Duan
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, 671000, PR China
| | - Siying Li
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, 671000, PR China
| | - Guangming Wang
- School of Clinical Medicine, Dali University, Dali, Yunnan, 671000, PR China.
- Center of Genetic Testing, The First Affiliated Hospital of Dali University, Dali, Yunnan, 671000, PR China.
| |
Collapse
|
2
|
Unadkat SV, Padhi BK, Bhongir AV, Gandhi AP, Shamim MA, Dahiya N, Satapathy P, Rustagi S, Khatib MN, Gaidhane A, Zahiruddin QS, Sah R, Serhan HA. Association between homocysteine and coronary artery disease-trend over time and across the regions: a systematic review and meta-analysis. Egypt Heart J 2024; 76:29. [PMID: 38409614 PMCID: PMC10897093 DOI: 10.1186/s43044-024-00460-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 02/21/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND The association of homocysteine with coronary artery disease (CAD) has been explored previously with mixed findings. The present Systematic Review and Meta-Analysis (SRMA) has assessed the pooled estimate of association between homocysteine (Hcy) and CAD, and its variation over the period and geography. METHODS Systematic literature search was done in PubMed, Scopus and Cochrane to identify the observational studies that have reported mean Hcy among cases (CAD) and control. The SRMA was registered in PROSPERO (ID-CRD42023387675). RESULTS Pooled standardized mean difference (SMD) of Hcy levels between the cases and controls was 0.73 (95% CI 0.55-0.91) from 59 studies. Heterogeneity was high (I2 94%). The highest SMD was found among the Asian studies (0.85 [95% CI 0.60-1.10]), while the European studies reported the lowest SMD between the cases and controls (0.32 [95% CI 0.18-0.46]). Meta-regression revealed that the strength of association was increasing over the years (Beta = 0.0227, p = 0.048). CONCLUSIONS Higher homocysteine levels might have a significant association with coronary artery diseases, but the certainty of evidence was rated low, owing to the observational nature of the studies, high heterogeneity, and publication bias. Within the population groups, Asian and African populations showed a greater strength of association than their European and American counterparts, and it also increased over the years.
Collapse
Affiliation(s)
- Sumit V Unadkat
- Department of Community Medicine, M. P. Shah Government Medical College, Jamnagar, Gujarat, India
| | - Bijaya K Padhi
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Aparna Varma Bhongir
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
| | - Aravind P Gandhi
- Department of Community Medicine, All India Institute of Medical Sciences, Nagpur, 441108, India.
| | - Muhammad Aaqib Shamim
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, 342005, India
- Global Center for Evidence Synthesis, Chandigarh, 160036, India
| | - Neelam Dahiya
- Department of Cardiology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Prakasini Satapathy
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248001, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, DMIHER, Wardha, India
| | - Abhay Gaidhane
- Jawaharlal Nehru Medical College, One Health Centre (COHERD), Datta Meghe Institute of Higher Education, Wardha, India
| | - Quazi Syed Zahiruddin
- Division of Evidence Synthesis, School of Epidemiology and Public Health and Research, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha, India
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Kathmandu, 46000, Nepal
- Department of Clinical Microbiology, DY Patil Medical College, Hospital and Research Centre, DY Patil Vidyapeeth, Pune, 411000, Maharashtra, India
| | | |
Collapse
|
3
|
Investigation of DHA-Induced Regulation of Redox Homeostasis in Retinal Pigment Epithelium Cells through the Combination of Metabolic Imaging and Molecular Biology. Antioxidants (Basel) 2022; 11:antiox11061072. [PMID: 35739970 PMCID: PMC9219962 DOI: 10.3390/antiox11061072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Diabetes-induced oxidative stress leads to the onset of vascular complications, which are major causes of disability and death in diabetic patients. Among these, diabetic retinopathy (DR) often arises from functional alterations of the blood-retinal barrier (BRB) due to damaging oxidative stress reactions in lipids, proteins, and DNA. This study aimed to investigate the impact of the ω3-polyunsaturated docosahexaenoic acid (DHA) on the regulation of redox homeostasis in the human retinal pigment epithelial (RPE) cell line (ARPE-19) under hyperglycemic-like conditions. The present results show that the treatment with DHA under high-glucose conditions activated erythroid 2-related factor Nrf2, which orchestrates the activation of cellular antioxidant pathways and ultimately inhibits apoptosis. This process was accompanied by a marked increase in the expression of NADH (Nicotinamide Adenine Dinucleotide plus Hydrogen) Quinone Oxidoreductase 1 (Nqo1), which is correlated with a contextual modulation and intracellular re-organization of the NAD+/NADH redox balance. This investigation of the mechanisms underlying the impairment induced by high levels of glucose on redox homeostasis of the BRB and the subsequent recovery provided by DHA provides both a powerful indicator for the detection of RPE cell impairment as well as a potential metabolic therapeutic target for the early intervention in its treatment.
Collapse
|
4
|
Zazueta C, Jimenez-Uribe AP, Pedraza-Chaverri J, Buelna-Chontal M. Genetic Variations on Redox Control in Cardiometabolic Diseases: The Role of Nrf2. Antioxidants (Basel) 2022; 11:antiox11030507. [PMID: 35326157 PMCID: PMC8944632 DOI: 10.3390/antiox11030507] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 02/07/2023] Open
Abstract
The transcription factor Nrf2 is a master regulator of multiple cytoprotective genes that maintain redox homeostasis and exert anti-inflammatory functions. The Nrf2-Keap1 signaling pathway is a paramount target of many cardioprotective strategies, because redox homeostasis is essential in cardiovascular health. Nrf2 gene variations, including single nucleotide polymorphisms (SNPs), are correlated with cardiometabolic diseases and drug responses. SNPs of Nrf2, KEAP1, and other related genes can impair the transcriptional activation or the activity of the resulting protein, exerting differential susceptibility to cardiometabolic disease progression and prevalence. Further understanding of the implications of Nrf2 polymorphisms on basic cellular processes involved in cardiometabolic diseases progression and prevalence will be helpful to establish more accurate protective strategies. This review provides insight into the association between the polymorphisms of Nrf2-related genes with cardiometabolic diseases. We also briefly describe that SNPs of Nrf2-related genes are potential modifiers of the pharmacokinetics that contribute to the inter-individual variability.
Collapse
Affiliation(s)
- Cecilia Zazueta
- Departmento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I.Ch., Mexico City 14080, Mexico;
| | - Alexis Paulina Jimenez-Uribe
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.P.J.-U.); (J.P.-C.)
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico; (A.P.J.-U.); (J.P.-C.)
| | - Mabel Buelna-Chontal
- Departmento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología, I.Ch., Mexico City 14080, Mexico;
- Correspondence:
| |
Collapse
|
5
|
S SP, K KE. Contribution of Glutathione-S-Transferases Polymorphism and Risk of Coronary Artery Diseases: A Meta-Analysis. Curr Aging Sci 2022; 15:282-292. [PMID: 35249517 DOI: 10.2174/1874609815666220304193925] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 11/15/2021] [Accepted: 01/07/2022] [Indexed: 12/06/2022]
Abstract
BACKGROUND Oxidative stress is one of the risk components in the development of coronary artery diseases (CAD) and polymorphism in major antioxidant genes like Glutathione-S-Transferases (GST) has been associated with the increased CAD susceptibility and severity. OBJECTIVE To get a precise evaluation and to update the association, a meta-analysis on GST (GSTM1, GSTT1 and GSTP1) polymorphism with CAD was performed. Moreover, the combined effect of GSTM1/GSTT1 null genotypes on CAD risk which is not yet studied so far but it has the highest risk of developing diseases. MATERIALS AND METHOD PubMed, Embase and Web of Science were systematically searched for eligible studies. Case-control studies with genotypic frequency, provide data to calculate odds ratio (OR) and in English language were selected. OR with 95% C.I was calculated and random effect model was used. NOS scale was used to asses the qulity of the included studies. RESULTS Meta-analysis indicated that the GSTM1 null genotype and GSTP1 (Ile105Val) polymorphism is significantly associated with CAD risk with a pooled OR-1.38, p=0.01 for GSTM1 and OR-1.19, p=0.04 for GSTP1. Dual null genotype of GSTM1-GSTT1 has highest risk for CAD development (OR-1.59, p=0.003) and there is no significant association for GSTT1 null genotype with CAD. In the subgroup analysis, GSTM1 showed the increased risk for Asians (OR-1.68, p=<0.01) and smokers (OR-1.98, p=<0.01). Publication bias was not observed. CONCLUSION The findings suggests that the GSTM1/GSTP1 polymorphism can be a predictive factor for CAD risk and a larger sample size is required further to confirm the association.
Collapse
Affiliation(s)
- Santhi Priya S
- Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute, Chettinad Academy of Research & Education, Kelambakkam-603 103, Chengalpattu Dt.,TN, India
| | - Kumar Ebenezar K
- Faculty of Allied Health Sciences, Chettinad Hospital & Research Institute, Chettinad Academy of Research & Education, Kelambakkam-603 103, Chengalpattu Dt.,TN, India
| |
Collapse
|
6
|
Xu LT, Wang T, Fang KL, Zhao Y, Wang XN, Ren DM, Shen T. The ethanol extract of flower buds of Tussilago farfara L. attenuates cigarette smoke-induced lung inflammation through regulating NLRP3 inflammasome, Nrf2, and NF-κB. JOURNAL OF ETHNOPHARMACOLOGY 2022; 283:114694. [PMID: 34601084 DOI: 10.1016/j.jep.2021.114694] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 09/13/2021] [Accepted: 09/27/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The flower buds of Tussilago farfara L. (Abbreviated as FTF) were widely used in traditional Chinese medicine (TCM) to treat respiratory diseases, including asthma, dry throat, great thirst, turbid saliva, stinky pus, and coughs caused by various causes. AIM OF STUDY The aim of study is to explore the efficiency of FTF in vitro and in vivo for the treatment of lung inflammation, and to illustrate the possible mechanisms of FTF in treating inflammation-related respiratory diseases targeting NOD-like receptor 3 (NLRP3) inflammasome, nuclear factor erythroid 2-related factor 2 (Nrf2), and nuclear transcription factor-κB (NF-κB). METHODS Lung inflammation model in vivo was induced by exposure of mice to cigarette smoke (CS) for two weeks. The levels of superoxide dismutase (SOD), malondialdehyde (MDA), inflammatory factors, and histology in lung tissues were investigated in presence or absence of ethanol extract of the flower buds of T. farfara L. (FTF-EtOH). In the cell-based models, nitric oxide (NO) assay, flow cytometry assay, enzyme-linked immunosorbent assay (Elisa), and glutathione (GSH) assay were used to explore the anti-inflammatory and anti-oxidant effects of FTF-EtOH. Possible anti-inflammatory mechanisms of FTF targeting NLRP3 inflammasome, Nrf2, and NF-κB have been determined using western blot, quantitative real-time reverse transcriptase-polymerase chain reaction (qRT-PCR), immunofluorescence assay, nuclear and cytoplasmic extraction, and ubiqutination assay. RESULTS FTF-EtOH suppressed CS-induced overproduction of inflammatory factors [e.g., tumor necrosis factor-α (TNF-α) and interleukin-1β (IL-1β)], and upregulation of the content of intracellular MDA in the lung homogenate of mice. In cell-based models, FTF-EtOH reduced the lipopolysaccharide (LPS)-induced overproduction of inflammatory factors, and attenuated the CS extract-induced overgeneration of reactive oxygen species (ROS). Furthermore, FTF-EtOH up-regulated Nrf2 and its downstream genes through enhancing the stability of Nrf2 protein, and inhibited the activation of NF-κB and NLRP3 inflammasome, which have been confirmed by detecting the protein levels in the mouse model. CONCLUSIONS FTF-EtOH effectively attenuated lung inflammation in vitro and in vivo. The protection of FTF-EtOH against inflammation was produced by activation of Nrf2 and inhibitions of NF-κB and NLRP3 inflammasome. These datas definitely support the ethnopharmacological use of FTF as an anti-inflammatory drug for treating respiratory diseases in TCM.
Collapse
Affiliation(s)
- Lin-Tao Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Tian Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Kai-Li Fang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Yu Zhao
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Xiao-Ning Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China
| | - Dong-Mei Ren
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, People's Republic of China.
| |
Collapse
|
7
|
Yan L, Xu D, Xiao Y, Dai M, Wang T, Zhuang X, Wu K. Genetic Polymorphism of NQO1 Gene is Associated with Susceptibility of Ischemic Stroke in Chinese Han Nationality. Neuropsychiatr Dis Treat 2022; 18:2211-2217. [PMID: 36196359 PMCID: PMC9527027 DOI: 10.2147/ndt.s379742] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
PURPOSE NAD(P)H: Quinone Oxidoreductase 1 gene (NQO1) polymorphism is associated with the risk of cardiovascular disease. This study was designed to investigate the relationship between NQO1 gene polymorphism and ischemic stroke susceptibility in Chinese Han nationality. PATIENTS AND METHODS One hundred and forty-one patients diagnosed with ischemic stroke and 139 matched control groups were recruited in this study. The polymorphism distribution of rsl800566 locus and rs10517 locus of NQO1 gene was genotyped via TaqMan assay, and the concentration of Oxidized low-density lipoprotein (ox-LDL) in the blood of the subjects was detected by enzyme linked immunosorbent assay (ELISA). The relationship between the polymorphism distribution and the susceptibility to ischemic stroke was evaluated. RESULTS The frequency distribution of the three genotypes of NQO1 rs1800566 between the case group and the control group was statistically significant, and cases carrying CT and TT genotype were less likely to suffer from ischemic stroke. Compared with individuals carrying T allele, C allele carriers have higher risk of ischemic stroke. However, there was no significant difference in frequency distribution among the three genotypes of NQO1 rs10517 between controls and patients. CONCLUSION The NQO1 rs1800566 C allele may be a novel marker associated with ischemic stroke susceptibility in Chinese Han population. Polymorphism of rsl800566 locus in NQO1 gene may be protective against ischemic stroke risk.
Collapse
Affiliation(s)
- Limin Yan
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Dedong Xu
- Department of Neurosurgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Ying Xiao
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Mingming Dai
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Ting Wang
- Department of Neurology, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Xinhong Zhuang
- Department of Nephrology, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| | - Kunliang Wu
- Department of Infectious Diseases, The Second Affiliated Hospital of Hainan Medical University, Haikou, People's Republic of China
| |
Collapse
|
8
|
Lee WS, Ham W, Kim J. Roles of NAD(P)H:quinone Oxidoreductase 1 in Diverse Diseases. Life (Basel) 2021; 11:life11121301. [PMID: 34947831 PMCID: PMC8703842 DOI: 10.3390/life11121301] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/15/2021] [Accepted: 11/18/2021] [Indexed: 01/07/2023] Open
Abstract
NAD(P)H:quinone oxidoreductase (NQO) is an antioxidant flavoprotein that catalyzes the reduction of highly reactive quinone metabolites by employing NAD(P)H as an electron donor. There are two NQO enzymes—NQO1 and NQO2—in mammalian systems. In particular, NQO1 exerts many biological activities, including antioxidant activities, anti-inflammatory effects, and interactions with tumor suppressors. Moreover, several recent studies have revealed the promising roles of NQO1 in protecting against cardiovascular damage and related diseases, such as dyslipidemia, atherosclerosis, insulin resistance, and metabolic syndrome. In this review, we discuss recent developments in the molecular regulation and biochemical properties of NQO1, and describe the potential beneficial roles of NQO1 in diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Wang-Soo Lee
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| | - Woojin Ham
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul 06974, Korea;
- Correspondence: (W.-S.L.); (J.K.); Tel.: +82-2-6299-1419 (W.-S.L.); +82-2-6299-1397 (J.K.)
| |
Collapse
|
9
|
Vasamsetti BMK, Chon K, Kim J, Oh JA, Yoon CY, Park HH. Transcriptome-Based Identification of Genes Responding to the Organophosphate Pesticide Phosmet in Danio rerio. Genes (Basel) 2021; 12:genes12111738. [PMID: 34828343 PMCID: PMC8624534 DOI: 10.3390/genes12111738] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 12/28/2022] Open
Abstract
Organophosphate pesticides (OPPs) are one of the most widely used insecticides. OPPs exert their neurotoxic effects by inhibiting acetylcholine esterase (AChE). Most of the gross developmental abnormalities observed in OPP-treated fish, on the other hand, may not be explained solely by AChE inhibition. To understand the overall molecular mechanisms involved in OPP toxicity, we used the zebrafish (ZF) model. We exposed ZF embryos to an OPP, phosmet, for 96 h, and then analyzed developmental abnormalities and performed whole transcriptome analysis. Phenotypic abnormalities, such as bradycardia, spine curvature, and growth retardation, were observed in phosmet-treated ZF (PTZF). Whole transcriptome analysis revealed 2190 differentially expressed genes (DEGs), with 822 and 1368 significantly up-and downregulated genes, respectively. System process and sensory and visual perception were among the top biological pathways affected by phosmet toxicity. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed significant enrichment of metabolic pathways, calcium signaling pathway, regulation of actin cytoskeleton, cardiac muscle contraction, drug metabolism-other enzymes, and phototransduction. Quantitative real-time PCR results of six DEGs agreed with the sequencing data expression profile trend. Our findings provide insights into the consequences of phosmet exposure in ZF, as well as an estimate of the potential risk of OPPs to off-target species.
Collapse
|
10
|
Role of Human NADPH Quinone Oxidoreductase (NQO1) in Oxygen-Mediated Cellular Injury and Oxidative DNA Damage in Human Pulmonary Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5544600. [PMID: 34691356 PMCID: PMC8536466 DOI: 10.1155/2021/5544600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 09/03/2021] [Indexed: 12/16/2022]
Abstract
Supplemental oxygen administration is frequently used in premature infants and adults with pulmonary insufficiency. NADPH quinone oxidoreductase (NQO1) protects cells from oxidative injury by decreasing reactive oxygen species (ROS). In this investigation, we tested the hypothesis that overexpression of NQO1 in BEAS-2B cells will mitigate cell injury and oxidative DNA damage caused by hyperoxia and that A-1221C single nucleotide polymorphism (SNP) in the NQO1 promoter would display altered susceptibility to hyperoxia-mediated toxicity. Using stable transfected BEAS-2B cells, we demonstrated that hyperoxia decreased cell viability in control cells (Ctr), but this effect was differentially mitigated in cells overexpressing NQO1 under the regulation of the CMV viral promoter, the wild-type NQO1 promoter (NQO1-NQO1), or the NQO1 promoter carrying the SNP. Interestingly, hyperoxia decreased the formation of bulky oxidative DNA adducts or 8-hydroxy-2′-deoxyguanosine (8-OHdG) in Ctr cells. qPCR studies showed that mRNA levels of CYP1A1 and NQO1 were inversely related to DNA adduct formation, suggesting the protective role of these enzymes against oxidative DNA injury. In SiRNA experiments entailing the NQO1-NQO1 promoter, hyperoxia caused decreased cell viability, and this effect was potentiated in cells treated with CYP1A1 siRNA. We also found that hyperoxia caused a marked induction of DNA repair genes DDB2 and XPC in Ctr cells, supporting the idea that hyperoxia in part caused attenuation of bulky oxidative DNA lesions by enhancing nucleotide excision repair (NER) pathways. In summary, our data support a protective role for human NQO1 against oxygen-mediated toxicity and oxidative DNA lesions in human pulmonary cells, and protection against toxicity was partially lost in SNP cells. Moreover, we also demonstrate a novel protective role for CYP1A1 in the attenuation of oxidative cells and DNA injury. Future studies on the mechanisms of attenuation of oxidative injury by NQO1 should help in developing novel approaches for the prevention/treatment of ARDS in humans.
Collapse
|
11
|
Sobha SP, Ebenezar K. Susceptibility of Glutathione--S-Transferase Polymorphism to CVD Development in Type 2 Diabetes Mellitus - A Review. Endocr Metab Immune Disord Drug Targets 2021; 22:225-234. [PMID: 34496736 DOI: 10.2174/1871530321666210908115222] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 12/06/2022]
Abstract
BACKGROUND Metabolic disorder affects normal homeostasis and can lead to the development of diseases. Diabetes mellitus is the most common metabolic disorder, and a cluster of metabolic conditions can lead to cardiovascular disease (CVD) development. Diabetes mellitus and CVD are closely related, with oxidative stress, playing a major role in the pathophysiology. Glutathione-S-Transferases (GST) potentially play an important role by reducing oxidative stress and is found to be the underlying pathophysiology in the development of diabetes, cardiovascular diseases (CVD), etc. Objectives: In this review, the role of GST genetic variant in the development of diabetes mellitus, CVD and diabetic vascular complications has been focused. RESULTS Based on the literature, it is evident that the GST can act as an important biochemical tool providing significant evidence regarding oxidative stress predominant in the development of diseases. Analysis of GST gene status, particularly detection of GSTM1 and GSTT1 null mutations and GSTP1 polymorphism, have clinical importance. CONCLUSION The analysis of GST polymorphism may help identify the people at risk and provide proper medical management. Genotyping of GST gene would be a helpful biomarker for early diagnosis of CVD development in DM and also in CVD cases. More studies focusing on the association of GST polymorphism with CVD development in diabetic patients will help us determine the pathophysiology better.
Collapse
Affiliation(s)
- Santhi Priya Sobha
- Natural Medicine and Molecular Physiology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam-603103, Chengalpattu Dt., TN. India
| | - Kumar Ebenezar
- Natural Medicine and Molecular Physiology Lab, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Kelambakkam-603103, Chengalpattu Dt., TN. India
| |
Collapse
|
12
|
Niksirat H, Siino V, Steinbach C, Levander F. High-Resolution Proteomic Profiling Shows Sexual Dimorphism in Zebrafish Heart-Associated Proteins. J Proteome Res 2021; 20:4075-4088. [PMID: 34185526 DOI: 10.1021/acs.jproteome.1c00387] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Understanding the molecular basis of sexual dimorphism in the cardiovascular system may contribute to the improvement of the outcome in biological, pharmacological, and toxicological studies as well as on the development of sex-based drugs and therapeutic approaches. Label-free protein quantification using high-resolution mass spectrometry was applied to detect sex-based proteome differences in the heart of zebrafish Danio rerio. Out of almost 3000 unique identified proteins in the heart, 79 showed significant abundance differences between male and female fish. The functional differences were mapped using enrichment analyses. Our results suggest that a large amount of materials needed for reproduction (e.g., sugars, lipids, proteins, etc.) may impose extra pressure on blood, vessels, and heart on their way toward the ovaries. In the present study, the female's heart shows a clear sexual dimorphism by changing abundance levels of numerous proteins, which could be a way to safely overcome material-induced elevated pressures. These proteins belong to the immune system, oxidative stress response, drug metabolization, detoxification, energy, metabolism, and so on. In conclusion, we showed that sex can induce dimorphism at the molecular level in nonsexual organs such as heart and must be considered as an important factor in cardiovascular research. Data are available via ProteomeXchange with identifier PXD023506.
Collapse
Affiliation(s)
- Hamid Niksirat
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Valentina Siino
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden
| | - Christoph Steinbach
- Faculty of Fisheries and Protection of Waters, CENAKVA, University of South Bohemia in České Budějovice, Vodňany, 370 05 České Budějovice, Czech Republic
| | - Fredrik Levander
- Department of Immunotechnology, Lund University, Lund 223 87, Sweden.,National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, Lund 223 87, Sweden
| |
Collapse
|
13
|
Zivkovic M, Bubic M, Kolakovic A, Dekleva M, Stankovic G, Stankovic A, Djuric T. The association of glutathione S-transferase T1 and M1 deletions with myocardial infarction. Free Radic Res 2021; 55:267-274. [PMID: 34003050 DOI: 10.1080/10715762.2021.1931166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Glutathione S-transferases (GSTs) are the family of enzymes involved in the second line of defense against oxidative stress (OS). The lack of GSTT1/GSTM1 enzyme quantity or activity, due to the presence of homozygous deletion compromises antioxidative defense resulting in OS. OS is the critical mechanism in the pathophysiology of atherosclerosis, coronary artery disease, and myocardial infarction (MI). The increase in reactive oxygen species together with the process of apoptosis plays a role in left ventricular remodeling (LVR) after MI. The associations of GSTT1 and GSTM1 gene polymorphisms with the risk of MI are inconsistent. The aim was to analyze the association of GSTT1/GSTM1 null genotypes with first MI and LVR 8 months after the MI. The study involved 330 controls and 438 consecutive patients with symptoms and signs of first MI. The subgroup of 150 MI patients was prospectively followed up for 6 months. Evidence of maladaptive LVR was obtained by 2D Doppler echocardiography 3-5 days and 6 months after the MI. A multiplex polymerase chain reaction was used to detect the deletion in GSTT1 and GSTM1 genes. GSTM1 null genotype was significantly and independently associated with first MI (adjusted OR = 1.45 95% CI 1.03-2.03, p = 0.03). Association of double null genotypes with maladaptive LVR in patients 6 months after the first MI was no longer significant after adjustment for factors that differed significantly between patients with and without maladaptive LVR. This study demonstrated the association of GSTM1 null genotypes with the risk of MI in the Serbian population.
Collapse
Affiliation(s)
- Maja Zivkovic
- Department of Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Maja Bubic
- Department of Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Kolakovic
- Department of Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milica Dekleva
- Department of Cardiology, University Clinical Center "Zvezdara", Belgrade, Serbia.,Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Goran Stankovic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia.,Cardiology Clinic, Clinical Center of Serbia, Belgrade, Serbia
| | - Aleksandra Stankovic
- Department of Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Tamara Djuric
- Department of Radiobiology and Molecular Genetics, "Vinca" Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
14
|
An updated meta-analysis showed smoking modify the association of GSTM1 null genotype on the risk of coronary heart disease. Biosci Rep 2021; 41:227694. [PMID: 33506866 PMCID: PMC7881159 DOI: 10.1042/bsr20200490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 01/14/2021] [Accepted: 01/22/2021] [Indexed: 12/06/2022] Open
Abstract
Background Oxidative stress is considered to be involved in the pathogenesis of coronary heart disease (CHD). Glutathione-S-transferase (GST) enzymes play important roles in antioxidant defenses and may influence CHD risk. The present meta-analysis was performed to investigate the link between glutathione S-transferase M1 (GSTM1) null genotype and CHD and to get a precise evaluation of interaction between GSTM1 null genotype and smoking by the case-only design. Methods PubMed and EMBASE databases were searched through 15 December 2020 to retrieve articles. Odds ratios (ORs) were pooled using either fixed-effects or random-effects models. Results Thirty-seven studies showed that GSTM1 null genotype was associated with risk of CHD in total population, Caucasians and Asians (for total population, OR = 1.38, 95% confidence interval (CI): 1.15, 1.65; for Caucasians, OR = 1.34, 95% CI: 1.04, 1.72; for Asians, OR = 1.40, 95% CI: 1.11, 1.77). After adjustment for heterogeneity, these relationships were still significant. After adjustment for heterogeneity, case-only analysis of 11 studies showed a positive multiplicative interaction between GSTM1 null genotype and smoking (ever smoking vs. never smoking) (OR = 1.27, 95% CI: 1.08, 1.50; I2 = 0%, P=0.553). Conclusions The overall results indicated that GSTM1 null genotype was associated with a higher risk of CHD, and the association may be affected by smoking status. This is the first meta-analysis to prove a positive effect of the interaction between GSTM1 null genotype and smoking status on the risk of CHD. Well-designed studies are needed to investigate the possible gene–gene or gene–environment interactions.
Collapse
|
15
|
Shukla H, Gaje G, Koucheki A, Lee HY, Sun X, Trush MA, Zhu H, Li YR, Jia Z. NADPH-quinone oxidoreductase-1 mediates Benzo-[a]-pyrene-1,6-quinone-induced cytotoxicity and reactive oxygen species production in human EA.hy926 endothelial cells. Toxicol Appl Pharmacol 2020; 404:115180. [PMID: 32739527 DOI: 10.1016/j.taap.2020.115180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 07/11/2020] [Accepted: 07/28/2020] [Indexed: 11/17/2022]
Abstract
Numerous studies conducted in the past have reported deaths in the human population due to cardiovascular diseases (CVD) on exposure to air particulate matter (APM). BP-1,6-quinone (BP-1,6-Q) is one of the significant components of APM. However, the mechanism(s) by which it can exert its toxicity in endothelial cells is not yet completely understood. NAD(P)H: quinone oxidoreductase-1 (NQO1) is expressed highly in myocardium and vasculature tissues of the heart and plays a vital role in maintaining vascular homeostasis. This study, demonstrated that BP-1,6-Q diminishes NQO1 enzyme activity in a dose-dependent manner in human EA.hy926 endothelial cells. The decrease in the NQO1 enzyme causes potentiation in BP-1,6-Q-mediated toxicity in EA.hy926 endothelial cells. The enhancement of NQO1 in endothelial cells showed cytoprotection against BP-1,6-Q-induced cellular toxicity, lipid, and protein damage suggesting an essential role of NQO1 in cytoprotection against BP-1,6-Q toxicity. Using various biochemical assays and genetic approaches, results from this study further demonstrated that NQO1 also plays a crucial role in BP-1,6-Q-induced production of reactive oxygen species (ROS). These findings will contribute to elucidating BP-1,6-Q mediated toxicity and its role in the development of atherosclerosis.
Collapse
Affiliation(s)
- Halley Shukla
- Department of Biology, University of North Carolina at Greensboro, NC, USA
| | - Gabriella Gaje
- Department of Biology, University of North Carolina at Greensboro, NC, USA
| | - Ashkon Koucheki
- Department of Biology, University of North Carolina at Greensboro, NC, USA
| | - Ho Young Lee
- Department of Biology, University of North Carolina at Greensboro, NC, USA
| | - Xiaolun Sun
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA
| | - Michael A Trush
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Hong Zhu
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, USA
| | - Y Robert Li
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, USA
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, NC, USA.
| |
Collapse
|
16
|
Sarutipaiboon I, Settasatian N, Komanasin N, Kukongwiriyapan U, Sawanyawisuth K, Intharaphet P, Senthong V, Settasatian C. Association of Genetic Variations in NRF2, NQO1, HMOX1, and MT with Severity of Coronary Artery Disease and Related Risk Factors. Cardiovasc Toxicol 2020; 20:176-189. [PMID: 31332605 DOI: 10.1007/s12012-019-09544-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
NRF2 is a transcription factor which, during oxidative stress, activates transcription of its target antioxidant genes. Polymorphisms in NRF2 and its target antioxidant genes: HMOX-1, NQO1, and MT, have been associated with cardiovascular diseases (CVDs) and diabetes in various ethnic groups, however, with variable results. The aim of this study was to investigate the association of NRF2, HMOX-1, NQO1, and MT gene polymorphisms with CVD risk factors in Thais. The study was conducted in two groups: group with high-risk for coronary artery disease (CAD) and health check-up group. Polymorphisms in NRF2 (rs6721961), NQO1 (rs1800566), MT1A (rs11640851), and HMOX-1 (rs2071746) were genotyped. Expressions of NRF2, HMOX-1, and NQO1 were also determined. In high-risk group, NRF2 rs6721961-TT was associated with CAD [OR (95% CI) 5.07 (1.42-18.10)] and severity of coronary atherosclerosis [Gensini score > 32, OR (95% CI) 4.31 (1.67-11.09)]; rs6721961 GT and TT revealed significant association with lower mRNA expression than GG (p = 0.021). NQO1 rs1800566 also revealed association with CAD, only in female. Combined effect of NQO1-rs1800566, HMOX1-rs2071746, and MT1A-rs11640851 was evaluated on the risks of DM and hypertension. With a combination of risk alleles as genetic risk score (GRS), the highest GRS (score 6) increased risk for hypertension, comparing with GRS 0-2 [OR (95% CI) 1.89 (1.02-3.49)]; group with score 5-6 revealed association with risk of DM [OR (95% CI) 1.481 (1.08-2.04)]. In conclusion, NRF2 rs6721961 associated with CAD and severity of coronary atherosclerosis. NQO1 rs1800566 also associated with CAD, only in female. Combined polymorphisms of three NRF2-regulated genes increased risk of DM and hypertension.
Collapse
Affiliation(s)
- Ingkarat Sarutipaiboon
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nongnuch Settasatian
- School of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nantarat Komanasin
- School of Medical Technology, Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Upa Kukongwiriyapan
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Kittisak Sawanyawisuth
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Phongsak Intharaphet
- Queen Sirikit Heart Center of the Northeast Hospital, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Vichai Senthong
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand.,Queen Sirikit Heart Center of the Northeast Hospital, Khon Kaen University, Khon Kaen, 40002, Thailand.,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Chatri Settasatian
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand. .,Cardiovascular Research Group, Khon Kaen University, Khon Kaen, 40002, Thailand.
| |
Collapse
|
17
|
Liu M, Guo J, Chen B, Xia J, Pu X, Zou X, Yang M, Sizhou Huang. The expression of zebrafish NAD(P)H:quinone oxidoreductase 1(nqo1) in adult organs and embryos. Gene Expr Patterns 2020; 38:119134. [PMID: 32889095 DOI: 10.1016/j.gep.2020.119134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 08/26/2020] [Indexed: 10/23/2022]
Abstract
NQO1, NAD(P)H: quinone oxidoreductase 1, was first identified in rat and its role has been extensively studied. Even the roles of NQO1 in the maintenance of physiological function and disease were largely addressed, whether the tissue specific functions of the NQO1 in organ development remains unknown. In the current study, we identified two NQO1 isoforms (isoform 1 and isoform 2) and examined the expression of nqo1 variants in adult zebrafish organs and embryos at different stages. In adult organs, RT-PCR result indicated that nqo1 variant 1 was mainly expressed in stomach and intestine, while nqo1 variant 2 was expressed in all organs investigated except for heart. Further, RT-PCR result showed that the nqo1 variant 1 and variant 2 were expressed at all the embryonic stages, but nqo1 variant 1 expression level was much lower than that of nqo1 variant 2. To specifically examine the expression pattern of these two different nqo1 variants, we did whole mount in situ hybridization and the results demonstrated that, both of them were maternally expressed at 8-cell stage, and they were all expressed ubiquitously at early stage. At 24 hpf, nqo1 variant 2 was mainly expressed in yolk cells, and slightly in head and eyes. At 48 hpf, nqo1 variant 2 was restricted in lateral line neuromasts. From 72 hpf to 144 hpf, nqo1 variant 2 was mainly restricted in branchial arch, liver, swimming bladder and lateral line neuromasts, while from 124 hpf to 192 hpf, nqo1 variant 2 only restricted in liver, and disappeared in lateral line neuromasts. On the contrary, at the late embryonic stage, nqo1 variant 1 was only expressed in liver and swimming bladder while not in branchial arch and lateral line neuromasts. In conclusion, we systematically analyzed the expression pattern of nqo1 variant 1 and variant 2 in zebrafish at different embryonic stages, and our data implied the possible role of nqo1 in regulating liver, branchial arch and lateral neuromasts development.
Collapse
Affiliation(s)
- Min Liu
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Jinping Guo
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Bingyu Chen
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Jiamin Xia
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Xiaohua Pu
- School of Biomedical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Xinyu Zou
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan, 610500, China
| | - Min Yang
- School of Laboratory Medicine, Chengdu Medical College, Chengdu, China
| | - Sizhou Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medical Sciences, Chengdu Medical College, Chengdu, Sichuan, 610500, China.
| |
Collapse
|
18
|
Rebai A, Chbili C, Ben Amor S, Hassine A, Ben Ammou S, Saguem S. Effects of glutathione S-transferase M1 and T1 deletions on Parkinson's disease risk among a North African population. Rev Neurol (Paris) 2020; 177:290-295. [PMID: 32359946 DOI: 10.1016/j.neurol.2020.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 02/05/2023]
Abstract
PURPOSE In this study, the effects of glutathione S-transferase polymorphisms Mu1 (GSTM1) and glutathione S-transferase polymorphisms Theta1 (GSTT1) on Parkinson's disease (PD) risk factor were evaluated in a Tunisian population. METHODS These polymorphisms were analyzed in 229 healthy Tunisian subjects and 64 Tunisian patients with PD, using a polymerase chain reaction (PCR). Statistical analysis was performed using SPSS 18.0. The relative associations between the GST genotypes and PD were assessed by calculating the odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS The study results demonstrated that the individuals with GSTM1 [OR=3.93, 95% CI: 1.98-7.92, P=10-6] and GSTT1 [OR=5.45, 95% CI: 2.90-10.30, p=10-6] were statistically associated with the risk of PD. A significant association was also found between the individuals with both GSTM1/T1 null genotypes and PD risk [OR=22.10, 95% CI: 6.99-73.75, P=10-6]. CONCLUSION These genotyping findings suggest that the absence of both GSTM1 and GSTT1 activity could be a contributory factor for the development of PD.
Collapse
Affiliation(s)
- A Rebai
- Metabolic Biophysics and Applied Pharmacology, Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse 4002, Tunisia; Faculty of Physical and Natural Mathematical Sciences of Tunis, University of Tunis El Manar, Tunis 2092, Tunisia.
| | - C Chbili
- Metabolic Biophysics and Applied Pharmacology, Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse 4002, Tunisia
| | - S Ben Amor
- Neurology Department of the University Hospital Center (UHC) Sahloul, Sousse University, Sousse 4002, Tunisia
| | - A Hassine
- Neurology Department of the University Hospital Center (UHC) Sahloul, Sousse University, Sousse 4002, Tunisia
| | - S Ben Ammou
- Neurology Department of the University Hospital Center (UHC) Sahloul, Sousse University, Sousse 4002, Tunisia
| | - S Saguem
- Metabolic Biophysics and Applied Pharmacology, Laboratory, Department of Biophysics, Faculty of Medicine Sousse, Sousse University, Sousse 4002, Tunisia
| |
Collapse
|
19
|
Chang VC, Cotterchio M, Bondy SJ, Kotsopoulos J. Iron intake, oxidative stress‐related genes and breast cancer risk. Int J Cancer 2020; 147:1354-1373. [DOI: 10.1002/ijc.32906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/21/2019] [Accepted: 01/20/2020] [Indexed: 01/05/2023]
Affiliation(s)
- Vicky C. Chang
- Dalla Lana School of Public Health University of Toronto Toronto ON Canada
- Prevention and Cancer Control Cancer Care Ontario, Ontario Health Toronto ON Canada
| | - Michelle Cotterchio
- Dalla Lana School of Public Health University of Toronto Toronto ON Canada
- Prevention and Cancer Control Cancer Care Ontario, Ontario Health Toronto ON Canada
| | - Susan J. Bondy
- Dalla Lana School of Public Health University of Toronto Toronto ON Canada
| | - Joanne Kotsopoulos
- Dalla Lana School of Public Health University of Toronto Toronto ON Canada
- Women's College Research Institute, Women's College Hospital Toronto ON Canada
| |
Collapse
|
20
|
Bhatti JS, Vijayvergiya R, Singh B, Bhatti GK. Genetic susceptibility of glutathione S-transferase genes (GSTM1/T1 and P1) to coronary artery disease in Asian Indians. Ann Hum Genet 2018; 82:448-456. [PMID: 30039864 DOI: 10.1111/ahg.12274] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 06/16/2018] [Accepted: 06/20/2018] [Indexed: 12/30/2022]
Abstract
Genetic polymorphisms in glutathione S-transferase (GST) genes may modulate the risk of cardiovascular diseases. The objective of present study was to investigate the potential association between the polymorphisms of GSTM1/T1 and P1 genes and their influence on diverse clinical parameters and oxidative stress biomarkers in coronary artery disease (CAD) patients in Asian Indians. The present study includes 562 angiographically confirmed CAD patients and 564 healthy control subjects from the north Indian population. Anthropometric and clinical measurements were performed for all the participants. The oxidative stress biomarkers including malondialdehyde and total antioxidant capacity were also measured. The genotyping of the GSTM1/T1 and P1 genes was performed using the multiplex-PCR and PCR-RFLP methods. The CAD patients exhibit significantly high values of waist circumference, waist-to-hip ratio, body fat (%), glucose, triglycerides, and very low-density lipoprotein, and reduced high-density lipoprotein levels compared to control subjects (P < 0.001). Malondialdehyde levels were significantly enhanced, and the total antioxidant capacity was reduced in CAD patients compared to controls (P < 0.001). However, no significant difference in body mass index and total cholesterol levels were observed in CAD patients and control subjects. The frequencies of the GSTM1 and GSTM1/T1 null genotypes in the CAD patients were significantly higher than the control subjects. In contrast, the GSTT1(-) genotype frequencies were significantly lower in CAD patients than the controls. Logistic regression analysis of the data revealed the null genotype of GSTM1 and the GG genotype of the GSTP1 (313A/G) gene were associated with an approximately twofold enhanced risk of developing CAD, whereas GSTT1(-) plays a defensive role against CAD development in north Indians. Upon stratification of data according to the genotypes of the GSTM1/T1 and P1 genes, we did not find significant a difference among the various metabolic traits in CAD patients and controls. Our results suggest that oxidative damage induced by lipid peroxidation with reduced antioxidant capacity and genetic variants in GST genes (GSTM1/T1 and P1) may modify the risk of CAD development in Asian Indian population.
Collapse
Affiliation(s)
- J S Bhatti
- Department of Biotechnology, Sri Guru Gobind Singh College, Chandigarh, India
| | - R Vijayvergiya
- Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - B Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India.,UGC Centre of Excellence in Nano Applications, UIPS building, Panjab University, Chandigarh, India
| | - G K Bhatti
- UGC Centre of Excellence in Nano Applications, UIPS building, Panjab University, Chandigarh, India
| |
Collapse
|
21
|
Chbili C, Hassine A, Fathallah N, Nouira M, Naija S, Ben Ammou S, Saguem S. Glutathione S-transferase M1 and T1 polymorphisms and the risk of mild hepatotoxicity induced by carbamazepine in a tunisian population study. BMC Neurol 2018. [PMID: 29523098 PMCID: PMC5844121 DOI: 10.1186/s12883-018-1013-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
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. 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. Null alleles were determined using a Polymerase Chain Reaction. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured by standard methods. RESULTS Our results showed that the frequencies of GSTM1 (-) null allele and GSTT1 null (-) allele were 74.4 and 17.8% respectively. The ALT and AST levels were elevated in 46 (35.7%) and 33 (25.6%) cases. The mean values of ALT and AST were approximately 1.32 and 3.61 times higher than the upper limit of normal levels, respectively. The values of ALT and AST were significantly higher in GSTM1 (-) allele than in GSTM1 (+) (p = 10-3.and 0.004, respectively). The level of ALT was significantly higher in combination of GSTM1 (-)/T1(-) than in combined GSTM1(-)/T1(+) and combined GSTM1(+)/T1(+) (p = 0.2 and 0.03, respectively), and that of AST was significantly higher in combination of GSTM1(-)/T1(-) and in combination of GSTM1(+)/T1(-) than in combination of GSTM1(+)/T1(+) (p = 10-3 and 10-3, respectively). CONCLUSIONS Our findings suggest that the GSTM1 (-) allele may be considered as a key factor for the development of carbamazepine-induced hepatotoxicity. Results related to GSTT (-) allele and elevation in AST levels should be considered with caution as AST may be elevated in other pathophysiological conditions.
Collapse
Affiliation(s)
- Chahra Chbili
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Avenue Mohamed Karoui, 4002, Sousse, Tunisia.
| | - Anis Hassine
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Avenue Mohamed Karoui, 4002, Sousse, Tunisia.,Neurology Department of Central Hospital University (CHU), Sousse University, 4002, Sousse, Tunisia
| | - Neila Fathallah
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Avenue Mohamed Karoui, 4002, Sousse, Tunisia
| | - Manel Nouira
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Avenue Mohamed Karoui, 4002, Sousse, Tunisia
| | - Salma Naija
- Neurology Department of Central Hospital University (CHU), Sousse University, 4002, Sousse, Tunisia
| | - Sofiene Ben Ammou
- Neurology Department of Central Hospital University (CHU), Sousse University, 4002, Sousse, Tunisia
| | - Saad Saguem
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Avenue Mohamed Karoui, 4002, Sousse, Tunisia
| |
Collapse
|
22
|
Boroumand M, Pourgholi L, Goodarzynejad H, Ziaee S, Hajhosseini-Talasaz A, Sotoudeh-Anvari M, Mandegary A. NQO1 C609T Polymorphism is Associated with Coronary Artery Disease in a Gender-Dependent Manner. Cardiovasc Toxicol 2017; 17:35-41. [PMID: 26690082 DOI: 10.1007/s12012-015-9353-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Findings on the association of NQO1 C609T polymorphism in the NQO1 gene and cardiovascular disease susceptibility are controversial. The objective of the current study was to examine the relationship between this polymorphism and the presence and severity of angiographically determined coronary artery disease (CAD). One-hundred and forty-five patients with newly diagnosed angiographically documented CAD (≥50 % luminal stenosis of any coronary vessel) as case group were compared to 139 controls (subjects with no luminal stenosis at coronary arteries). The presence of C609T polymorphism was analyzed using polymerase chain reaction-based restriction fragment length polymorphism. Among total population, those with combined CT/TT (T allele carrier) genotype showed a trend toward lower odds of CAD compared to those with CC (wild type) genotype, but it did not reach a statistically significant level (p = 0.061). When data were analyzed separately for men or women, CT + TT group as compared to CC genotype was associated with decreased odds of CAD in women (adjusted OR 0.4, 95 % CI 0.2-0.9; p = 0.043), but not in men (adjusted OR 0.8, 95 % CI 0.3-1.9; p = 0.612). The C609T polymorphism within NQO1 is independently associated with CAD in women, but no association was observed in whole study population or in men.
Collapse
Affiliation(s)
- Mohammadali Boroumand
- Department of Clinical Pathology and Laboratory Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, P.O. Box 1411713138, Iran
| | - Leyla Pourgholi
- Department of Clinical Pathology and Laboratory Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, P.O. Box 1411713138, Iran.,Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, P.O. Box 7616911319, Iran
| | - Hamidreza Goodarzynejad
- Department of Cardiac Research, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, P.O. Box 1411713138, Iran
| | - Shayan Ziaee
- Department of Clinical Pathology and Laboratory Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, P.O. Box 1411713138, Iran
| | - Azita Hajhosseini-Talasaz
- Department of Clinical Pharmacy, School of Pharmacy, Tehran University of Medical Sciences, Tehran, P.O. Box 141556451, Iran
| | - Maryam Sotoudeh-Anvari
- Department of Clinical Pathology and Laboratory Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, P.O. Box 1411713138, Iran
| | - Ali Mandegary
- Department of Pharmacology and Toxicology, School of Pharmacy, Kerman University of Medical Sciences, Kerman, P.O. Box 7616911319, Iran. .,Gastroenterology and Hepatology Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, P.O. Box 7616911319, Iran.
| |
Collapse
|
23
|
Sharma M, Mehndiratta M, Gupta S, Kalra OP, Shukla R, Gambhir JK. Genetic association of NAD(P)H quinone oxidoreductase (NQO1*2) polymorphism with NQO1 levels and risk of diabetic nephropathy. Biol Chem 2017; 397:725-30. [PMID: 27078674 DOI: 10.1515/hsz-2016-0135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 04/06/2016] [Indexed: 01/23/2023]
Abstract
NAD(P)H quinone oxidoreductase 1 (NQO1) catalyzes reactions having a cyto-protective effect against redox cycling and oxidative stress. A single base polymorphism (C/T) at nucleotide 609 of the NQO1 gene impairs the stability and function of its protein. Its role in the development of diabetic nephropathy (DN) has not been deciphered. Therefore, this study aimed to evaluate the association of NQO1*2 (rs1800566) polymorphism with plasma NQO1 levels and DN. This study screened 600 participants including healthy controls (HC), type 2 diabetes mellitus without complications (T2DM) and diabetic nephropathy (DN): 200 each for studying NQO1*2 gene polymorphism using the PCR-RFLP. Plasma NQO1 levels were measured by ELISA. Analysis of variance and logistic regression were used to evaluate the association of NQO1 polymorphism with plasma NQO1 levels and DN. The allelic frequencies of NQO1*1/NQO1*2 were 0.88/0.12 in HC, 0.765/0.235 in T2DM and 0.65/0.35 in DN. Carriers of the NQO1*2 allele had significantly lower plasma NQO1 levels (p<0.05) and revealed higher risk towards the development of DN (OR=1.717, p=0.010). NQO1*2 SNP is a functional polymorphism having a significant effect on NQO1 levels. Our results indicate that NQO1*2 genotype may increase susceptibility to DN in north Indian subjects with T2DM.
Collapse
|
24
|
Chbili C, Elouaer A, Fathallah N, Nouira M, Jrad BBH, Gaha L, Saguem S. Effects of glutathione S-transferase M1 andT1 deletions on bipolar disorder risk among a Tunisian population. Gene 2017; 607:31-35. [DOI: 10.1016/j.gene.2016.12.035] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 11/14/2016] [Accepted: 12/29/2016] [Indexed: 01/11/2023]
|
25
|
Türkanoğlu Özçelik A, Can Demirdöğen B, Demirkaya Ş, Adalı O. Association of cytochrome P4502E1 and NAD(P)H:quinone oxidoreductase 1 genetic polymorphisms with susceptibility to large artery atherosclerotic ischemic stroke: a case–control study in the Turkish population. Neurol Sci 2017; 38:1077-1085. [DOI: 10.1007/s10072-017-2930-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 03/20/2017] [Indexed: 01/08/2023]
|
26
|
Glutathione S-Transferase T1 (GSTT1) Null Polymorphism, Smoking, and Their Interaction in Coronary Heart Disease: A Comprehensive Meta-Analysis. Heart Lung Circ 2016; 26:362-370. [PMID: 27686690 DOI: 10.1016/j.hlc.2016.07.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/30/2016] [Accepted: 07/07/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND The association between glutathione S-transferase T1 (GSTT1) null polymorphism and coronary heart disease (CHD) is inconsistent among studies, and data on the GSTT1 null genotype-smoking interplay in CHD is lacking. We conducted this meta-analysis to investigate the relationship between GSTT1 null polymorphism and CHD and to assess the potential interaction between GSTT1 null genotype and smoking. METHODS PubMed and EMBASE databases were searched up to 27 January 2016 using the appropriate terms. Odds ratios were pooled using either fixed-effects or random-effects models. RESULTS Twenty-nine articles including 31 studies with 15,004 cases and 35,597 controls were eligible. The random-effects model showed that the GSTT1 null genotype was associated with increased CHD risk (OR=1.213, 95%CI: 1.004-1.467; I2=90.4%). After excluding 10 studies detected by Galbraith plot, the fixed effects summary estimate also showed an increased risk of CHD (OR=1.14, 95% CI: 1.06-1.22; I2=27.7%). A case-only analysis including eight studies showed a statistically significant positive interaction between GSTT1 null polymorphism and smoking status on CHD (OR=1.34, 95% CI: 1.09-1.64; I2=0%). Sensitivity analyses further supported the associations. No publication bias was observed. CONCLUSIONS This meta-analysis suggests that GSTT1 null polymorphism is associated with the risk of CHD. To our knowledge, this is the first meta-analysis to prove a positive effect of the interaction between GSTT1 null genotype and smoking status on the risk of CHD. Future studies with detailed individual information are needed to confirm our findings.
Collapse
|
27
|
Rafiee L, Shokouh P, Roohafza H, Mansourian M, Javanmard SH. Association of glutathione S-transferases M1 and T1 gene polymorphisms with the risk of metabolic syndrome in an Iranian population. Adv Biomed Res 2016; 5:63. [PMID: 27135032 PMCID: PMC4832886 DOI: 10.4103/2277-9175.179185] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 02/23/2015] [Indexed: 01/01/2023] Open
Abstract
Background: Glutathione S-transferases (GSTs) are important factors in cell sensitivity to oxidative stress and susceptibility to cardiometabolic disorders. We aimed to investigate the GSTM1 and T1 gene polymorphisms, as well as their interactions in metabolic syndrome (MetS) patients and healthy individuals in an Iranian population. Materials and Methods: The study sample comprised of 220 healthy individuals (mean age: 41.9 – 15.1 years) and 165 MetS patients (mean age: 49.7 – 11.5 years). The diagnostic criteria for MetS were defined following the criteria provided by the modified National Cholesterol Education Program Adult Treatment Panel III. Genotyping of GSTM1 and T1 genes were performed using polymerase chain reaction. Results: Our analyses have shown that neither GSTM1 (odds ratio [OR] =0.89, 95% confidence interval [CI]: 0.59 – 1.33, P = 0.57) nor GSTT1 (OR = 1.26, 95% CI: 0.76 – 2.02, P = 0.38) null genotypes were associated with increased risk. Moreover, no significant differences were observed between various combinations of GST genotypes. Conclusion: Contrary to our primary hypothesis, what we found disaffirms any kind of association between GSTM1 and T1 polymorphisms and the risk of MetS. However, being the first polymorphism study of GSTs in MetS patients, further studies are required to confirm our results in other populations.
Collapse
Affiliation(s)
- Laleh Rafiee
- Applied Physiology Research Centre, Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Pedram Shokouh
- Cardiovascular Research Centre, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamidreza Roohafza
- Cardiovascular Research Centre, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marjan Mansourian
- Department of Biostatistics and Epidemiology, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
28
|
Rougée LRA, Riches Z, Berman JM, Collier AC. The Ontogeny and Population Variability of Human Hepatic NADPH Dehydrogenase Quinone Oxido-Reductase 1 (NQO1). ACTA ACUST UNITED AC 2016; 44:967-74. [PMID: 26856346 DOI: 10.1124/dmd.115.068650] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 02/05/2016] [Indexed: 01/16/2023]
Abstract
The NADPH dehydrogenase quinone oxido-reductase 1 (NQO1) enzyme is an antioxidant and metabolic enzyme that performs two electron reduction of quinones and other chemicals. Based on the physiologic role(s) of NQO1, we hypothesized that expression and activity of this enzyme would vary with age and other demographic variables. Cytosols from 117 archived human livers were investigated for changes in NQO1 with age, sex, obesity, and ethnicity. Protein expression but not activity of NQO1 was weakly negatively correlated with age (Spearman r = -0.2, P = 0.03). No sex differences were observed for either protein expression or activity and for ethnicity; Caucasians had greater NQO1 activity than Asians (P < 0.05). Overweight children had statistically significantly higher NQO1 activity as compared with ideal weight children (P < 0.05) although this difference was not observed in adults. These findings establish that NQO1 is approximately as active in children as adults. However, modeled NQO1 clearance (both allometric and physiologically based pharmacokinetics) predicted maturation at 23 to 26 years. This is almost certainly an overestimate, with error in the model resulting from a small sample size and inability to scale for age-related changes in hepatic cellularity and/or cytosolic protein content, and indicates a delay in reaching maximum clearance through the NQO1 pathway that is affected by physiologic development as much, or more than, biochemical development. Obesity may increase hepatic NQO1 activity in children, which is likely a protective mechanism in oxidative stress, but may also have significant implications for drug and chemical disposition in obese children.
Collapse
Affiliation(s)
- Luc R A Rougée
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Zoe Riches
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Jacob M Berman
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| | - Abby C Collier
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii (L.R.A.R., A.C.C.); Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada (Z.R., J.M.B., A.C.C.)
| |
Collapse
|
29
|
Glutathionyl systems and metabolic dysfunction in obesity. Nutr Rev 2015; 73:858-68. [DOI: 10.1093/nutrit/nuv042] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 04/18/2015] [Indexed: 12/18/2022] Open
|
30
|
Association of HMOX1 and NQO1 Polymorphisms with Metabolic Syndrome Components. PLoS One 2015; 10:e0123313. [PMID: 25933176 PMCID: PMC4416764 DOI: 10.1371/journal.pone.0123313] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/06/2015] [Indexed: 12/23/2022] Open
Abstract
Metabolic syndrome (MetS) is among the most important public health problems worldwide, and is recognized as a major risk factor for various illnesses, including type 2 diabetes mellitus, obesity, and cardiovascular diseases. Recently, oxidative stress has been suggested as part of MetS aetiology. The heme oxygenase 1 (HMOX1) and NADH:quinone oxidoreductase 1 (NQO1) genes are crucial mediators of cellular defence against oxidative stress. In the present study, we analysed the associations of HMOX1 (GT)n and NQO1 C609T polymorphisms with MetS and its components. Our study population comprised 735 Mexican Mestizos unrelated volunteers recruited from different tertiary health institutions from Mexico City. In order to know the HMOX1 (GT)n and NQO1 C609T allele frequencies in Amerindians, we included a population of 241 Amerindian native speakers. Their clinical and demographic data were recorded. The HMOX1 (GT)n polymorphism was genotyped using PCR and fluorescence technology. NQO1 C609T polymorphism genotyping was performed using TaqMan probes. Short allele (<25 GT repeats) of the HMOX1 polymorphism was associated with high systolic and diastolic blood pressure, and the T allele of the NQO1 C609T polymorphism was associated with increased triglyceride levels and decreased HDL-c levels, but only in individuals with MetS. This is the first study to analyse the association between MetS and genes involved in oxidative stress among Mexican Mestizos. Our data suggest that polymorphisms of HMOX1 and NQO1 genes are associated with a high risk of metabolic disorders, including high systolic and diastolic blood pressure, hypertriglyceridemia, and low HDL-c levels in Mexican Mestizo individuals.
Collapse
|
31
|
Yuan L, Ma W, Liu J, Meng L, Liu J, Li S, Han J, Liu Q, Feng L, Wang C, Xiao R. Effects of GSTM1/GSTT1 gene polymorphism and fruit & vegetable consumption on antioxidant biomarkers and cognitive function in the elderly: a community based cross-sectional study. PLoS One 2014; 9:e113588. [PMID: 25420021 PMCID: PMC4242636 DOI: 10.1371/journal.pone.0113588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 10/27/2014] [Indexed: 01/01/2023] Open
Abstract
Background It was reported that Glutathione S-transferase (GST) gene polymorphism and fruit and vegetable (FV) intake were associated with body antioxidant capacity. The oxidative/anti-oxidative imbalance played an important role in the pathogenesis of AD. However, the association of GST genotype, dietary FV consumption with body antioxidant biomarkers and cognitive function in the elderly is not clear. Objective The aim of the present study was to determine the association of GST genotype, and dietary FV intake, with antioxidant biomarkers and cognitive function in the elderly. Methods Food frequency questionnaire was used to collect data of dietary FV intakes in 504 community dwelling elderly aged from 55 to 75 years old. GSTM1 and GSTT1 genotypes were determined by using multiple-PCR method. Plasma and erythrocyte antioxidant biomarkers were measured. Cognitive function was measured by using Montreal Cognitive Assessment. Statistical analysis was applied for exploring the association of GST genotype and FV intake with antioxidant biomarkers level and cognitive function in the elderly. Results Individual GSTM1 or GSTT1 gene deletion affects body antioxidant biomarkers levels, including erythrocyte GST activity, plasma total antioxidant capacity, and glutathione levels. GSTM1and/or GSTT1 gene deletion have no effects on cognitive function in the surveyed participants. The effect of GST genotype on antioxidant biomarkers are FV intake dependent. There is interaction of FV intake and GST genotype on cognitive function in the elderly. Conclusion GST genotype or daily FV consumption impact body antioxidant biomarkers, but not cognitive function in the elderly. There were combined effects of GST genotype and FV consumption on cognitive function in the elderly population. Large scale perspective population study is required to explore the association of GST genetic polymorphism, FV consumption and antioxidant biomarkers and cognitive function in the elderly.
Collapse
Affiliation(s)
- Linhong Yuan
- School of Public Health, Capital Medical University, Beijing, P.R.China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, P.R. China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Jinmeng Liu
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Liping Meng
- Institute of Nutrition and Food Safety, Chinese Center for Disease Control and Prevention, Beijing, P.R. China
| | - Jixia Liu
- Nanyuan Community Health Service Center of Fengtai District, Beijing, P.R. China
| | - Shuang Li
- Nanyuan Community Health Service Center of Fengtai District, Beijing, P.R. China
| | - Jing Han
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Quanri Liu
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Lingli Feng
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Chao Wang
- School of Public Health, Capital Medical University, Beijing, P.R.China
| | - Rong Xiao
- School of Public Health, Capital Medical University, Beijing, P.R.China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, P.R. China
- * E-mail:
| |
Collapse
|
32
|
Nevzorova VA, Panchenko EA, Isaeva MP. DELETIONAL POLYMORPHISM OF GLUTATHIONE TRANSFERASE T1 AND M1 GENES IN PATIENTS WITH MYOCARDIAL INFARCTION AND METABOLIC SYNDROME. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2014. [DOI: 10.15829/1728-8800-2014-3-47-52] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Aim. To study associations of deletional polymorphism of genes GSTM1 and GSTT1 with myocardial infarction (MI) development on the background of metabolic syndrome (MS).Material and methods. Totally 86 patients with STEMI were included (from those 45 had signs of MS) and 30 healthy persons as control. By polymerase chain reaction the data on deletional polymorphism was obtained on the genes coding glutathiontransferase GSTM1 and GSTT1 in both groups.Results. The significant increase of prevalence of genotype GSTT10/0 is ascertained in patients with MI and without MS; presence of the “null” genotype increases relative risk of infarction in both patients with and without MS. In the first group (without MS) presence of genotype GSTT10/0 leads to increase of MI risk irrespective of smoking status, though in MS patients — only in those who smokes.Conclusion. Diagnostic of homozygous deletional mutation GSTT1 can be used for individual prognosis of MI risk in patients with MI risk factors — smoking and MS.
Collapse
Affiliation(s)
- V. A. Nevzorova
- SBEI HPE “Pacific Ocean State Medical University” of the Ministry of Health
| | - E. A. Panchenko
- SBEI HPE “Pacific Ocean State Medical University” of the Ministry of Health
| | - M. P. Isaeva
- FSBSI “Pacific Ocean Institute for Bio-organic Chemistry n. a. Elyakov G. B.” of RAS. Vladivostok, Russia
| |
Collapse
|
33
|
Chbili C, B'chir F, Ben Fredj M, Saguem BN, Ben Amor S, Ben Ammou S, Saguem S. Effects of glutathione S-transferase M1 and T1 deletions on epilepsy risk among a Tunisian population. Epilepsy Res 2014; 108:1168-73. [PMID: 24970787 DOI: 10.1016/j.eplepsyres.2014.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/10/2014] [Accepted: 05/17/2014] [Indexed: 01/11/2023]
Abstract
Glutathione-S-transferases enzymes are involved in the detoxification of several endogenous and exogenous substances. In this present study, we evaluated the effects of two glutathione-S-transferase polymorphisms, (GSTM1 and GSTT1) on epilepsy risk susceptibility in a Tunisian population. These polymorphisms were analyzed in 229 healthy subjects and 98 patients with epilepsy, using a polymerase chain reaction (PCR). Odds ratio (ORs) was used for analyzing results. The study results demonstrated that individuals with the GSTM1 null genotype were at an increased risk of developing epilepsy [OR=3.80, 95% confidence interval (CI) (2.15-4.78)], whereas no significant effects were observed between individuals with GSTT1 null genotype and epilepsy risk [OR=1.15, 95% CI (0.62-2.12)]. These genotyping finding revealed that the absence of GSTM1 activity could be contributor factor for the development of epilepsy disease.
Collapse
Affiliation(s)
- Chahra Chbili
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Sousse 4002, Tunisia.
| | - Fatma B'chir
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Sousse 4002, Tunisia; Laboratory of Natural Substances, National Institute of Research and Physical-Chemical Analysis (INRAP), Technopole Sidi Thabet 2020, Tunisia
| | - Maha Ben Fredj
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Sousse 4002, Tunisia
| | - Bochra-Nourhène Saguem
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Sousse 4002, Tunisia
| | - Sana Ben Amor
- Neurology Department of Central Hospital University (CHU), Sousse University, Sousse 4002, Tunisia
| | - Sofiene Ben Ammou
- Neurology Department of Central Hospital University (CHU), Sousse University, Sousse 4002, Tunisia
| | - Saad Saguem
- Metabolic Biophysics, Professional Toxicology and Applied Environmental Laboratory, Department of Biophysics, Medicine Faculty of Sousse, Sousse University, Sousse 4002, Tunisia.
| |
Collapse
|
34
|
Zhou D, Hu W, Wang Q, Jin Y. Glutathione S-transferase M1 polymorphism and coronary heart disease susceptibility: a meta-analysis involving 47,596 subjects. Heart Lung Circ 2014; 23:578-85. [PMID: 24525147 DOI: 10.1016/j.hlc.2014.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2013] [Revised: 12/26/2013] [Accepted: 01/11/2014] [Indexed: 01/08/2023]
Abstract
BACKGROUND Many studies have investigated the association between glutathione S-transferase M1 (GSTM1) null genotype and the risk of coronary heart disease (CHD). However, the effect of the GSTM1 null genotype on CHD is still unclear because of apparent inconsistencies among those studies. A meta-analysis was performed to characterise the relationship more accurately. METHODS Pubmed, Embase, and Web of Science were searched. We estimated the summary odds ratio (OR) with a 95% confidence interval (95% CI) to assess the association. RESULTS Up to 26 case-control studies with 13,929 CHD cases and 33,667 control cases were included into this meta-analysis. Meta-analysis of the 26 studies showed that GSTM1 null genotype was associated with the risk of CHD (random effects OR=1.35, 95% CI 1.00 to 1.83). After adjustment for heterogeneity, meta-analysis showed that GSTM1 null genotype was not associated with increased risk of CHD in the total population (fixed effects OR=1.01, 95% CI 0.95 to 1.07). In the subgroup analysis by ethnicity, increased risks were not found for either Caucasians (OR=1.36, 95% CI=0.96-1.92) or Asians (OR=1.28, 95% CI=0.91-1.80). When stratified by smoking status, in the subgroup of smokers, GSTM1 null genotype was significantly associated with increased CHD risk (random effects OR=1.64, 95% CI 1.02 to 2.64). No evidence of publication bias was observed. CONCLUSION In conclusion, this meta-analysis suggested that there is overall lack of association between GSTM1 genotypes and CHD risk, however, GSTM1 null genotype when combining with smoking history may contribute to CHD susceptibility.
Collapse
Affiliation(s)
- Donghui Zhou
- Department of Cardiology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Weina Hu
- Department of Cardiology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Qi Wang
- Department of Cardiology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China
| | - Yuanzhe Jin
- Department of Cardiology, the Fourth Affiliated Hospital of China Medical University, Shenyang 110032, China.
| |
Collapse
|
35
|
Dai D, Wang Y, Wang L, Li J, Zhou H, Ma Q, Zhou X, Pan J, Pan G, Chen C, Xu L, Ru P, Wang H, Zhu S, Lv Y, Xu L, Ye M, Duan S. Association of four GSTs gene polymorphisms with Parkinson disease: A meta-analysis. ACTA ACUST UNITED AC 2014. [DOI: 10.4236/abb.2014.52014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Schreiber R, Mill JG, Krieger JE, Pereira AC, Nadruz W. Association between glutathione S-transferase M1 polymorphism and urinary sodium excretion in a Brazilian population. Am J Hypertens 2013; 26:1024-9. [PMID: 23690164 DOI: 10.1093/ajh/hpt066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Null genotypes of glutathione S-transferase (GST) exhibit the absence of enzymatic activity and are associated with increased cardiovascular risk. Recent reports have related both lower and higher urinary sodium excretion (USE) to higher cardiovascular risk. Here we investigate the impact of GSTM1 and GSTT1-null polymorphisms on USE in a Brazilian population. METHODS We cross-sectionally evaluated 1,308 subjects from the city of Vitoria, Brazil, based on clinical history, physical examination, anthropometry, analysis of laboratory parameters, measurement of USE, and GST polymorphisms genotyping. RESULTS The frequency of GST M1, T1, and double-deletion polymorphisms was 51%, 22%, and 11%, respectively. Individuals with the GSTM1-null genotype had lower USE than those with the non-null genotype (92.1±52.3 vs. 102.8 ± 6 0.7 mEq/12h; P < 0.001). Linear regression analysis adjusted for confounding factors revealed that the GSTM1-null genotype was independently associated with USE (P = 0.001). In addition, diastolic blood pressure and triglyceride levels were higher in GSTM1-null individuals than in non-null individuals in the highest tertile of USE. Finally, the presence of GSTT1-null or double-deleted genotypes did not influence USE or affect the interactions between USE and the variables studied. CONCLUSIONS Deletion of GSTM1 was associated with low USE and modulated the interaction between sodium intake and blood pressure in Brazilian subjects. These novel findings may provide a new unexplored link between sodium regulation and GST homeostasis.
Collapse
Affiliation(s)
- Roberto Schreiber
- Department of Internal Medicine, University of Campinas, Campinas, Brazil
| | | | | | | | | |
Collapse
|
37
|
Analysis of glutathione S-transferase genes polymorphisms and the risk of schizophrenia in a sample of Iranian population. ACTA ACUST UNITED AC 2012; 7:199-203. [PMID: 22874804 DOI: 10.1017/s1740925x12000130] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Glutathione S-transferases (GSTs) are major intracellular antioxidants, which, impaired in their function, are involved in the progress of schizophrenia (SCZ). The aim of this case-control study was to investigate the association between the polymorphism of glutathione S-transferases M1 (GSTM1), T1 (GSTT1), the glutathione S-transferase P1 gene (GSTP1) and SCZ. We isolated genomic DNA from peripheral blood of 93 individuals with SCZ and 99 healthy control subjects' genotypes analyzing them for GSTM1, GSTT1 and GSTP1 using polymerase chain reaction. 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). The AG and combined AG + GG genotypes of GSTP1 increased the risk of SCZ (OR = 1.83; 95% CI: 0.94-3.75 and OR = 1.71; 95% CI: 0.92-3.19, respectively). The genotypes of GSTT/NULL, NULL/GSTM and NULL/NULL increased the risk of SCZ (OR = 2.05; 95% CI: 0.9-4.74; OR = 2.0; 95% CI: 1.68-2.31; and OR = 1.8; 95% CI: 0.57-2.46, respectively). The present study supports previous data that suggest that impairment in the function of GSTs genes may increase the risk of SCZ.
Collapse
|
38
|
GSTT1 null genotype contributes to coronary heart disease risk: a meta-analysis. Mol Biol Rep 2012; 39:8571-9. [DOI: 10.1007/s11033-012-1691-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 06/06/2012] [Indexed: 10/28/2022]
|
39
|
Conklin DJ, Bhatnagar A. Are glutathione S-transferase null genotypes "null and void" of risk for ischemic vascular disease? ACTA ACUST UNITED AC 2012; 4:339-41. [PMID: 21846869 DOI: 10.1161/circgenetics.111.960526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Taspinar M, Aydos S, Sakiragaoglu O, Duzen IV, Yalcinkaya A, Oztuna D, Bardakci H, Tutar E, Sunguroglu A. Impact of Genetic Variations of theCYP1A1, GSTT1, andGSTM1Genes on the Risk of Coronary Artery Disease. DNA Cell Biol 2012; 31:211-8. [DOI: 10.1089/dna.2011.1252] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Mehmet Taspinar
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Sena Aydos
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Onur Sakiragaoglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Irfan Veysel Duzen
- Department of Cardiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Adnan Yalcinkaya
- Department of Cardiovascular Surgery, Turkiye Yuksek Ihtisas Hospital, Ankara, Turkey
| | - Derya Oztuna
- Department of Biostatistics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Hasmet Bardakci
- Department of Cardiovascular Surgery, Turkiye Yuksek Ihtisas Hospital, Ankara, Turkey
| | - Eralp Tutar
- Department of Cardiology, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Asuman Sunguroglu
- Department of Medical Biology, Faculty of Medicine, Ankara University, Ankara, Turkey
| |
Collapse
|
41
|
Genetic association of Glutathione peroxidase-1 (GPx-1) and NAD(P)H:Quinone Oxidoreductase 1(NQO1) variants and their association of CAD in patients with type-2 diabetes. Mol Cell Biochem 2011; 361:143-50. [DOI: 10.1007/s11010-011-1098-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/28/2011] [Indexed: 10/17/2022]
|
42
|
The role of glutathione S-transferase M1 and T1 gene polymorphisms and fruit and vegetable consumption in antioxidant parameters in healthy subjects. Br J Nutr 2011; 107:928-33. [DOI: 10.1017/s0007114511003746] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The correlation of glutathione S-transferase (GST) M1/T1 genetic polymorphisms with oxidative stress-related chronic diseases was proved recently. The aim of the present study was to investigate the association of GSTM1/T1 genetic polymorphisms with antioxidant biomarkers and consumption of fruits and vegetables (F&V) in healthy subjects. In this study, for conducting a 3 d dietary survey, 190 healthy adults were recruited. After DNA extraction, a multiple PCR method was used for GSTM1/T1 genotyping. A spectrophotometer method was applied for the determination of plasma total antioxidant capacity (T-AOC), vitamin C level and erythrocyte GST enzyme activity. A general linear model was used to compare the mean values of antioxidant parameters for different GSTM1/T1 genotypes and consumption of F&V. Polymorphisms of GSTM1/T1 had no effects on plasma T-AOC and vitamin C levels. Deletion of the GSTM1 gene decreased the erythrocyte GST activity. There was correlation between plasma T-AOC and consumption of F&V in the GSTM1− or GSTT1+ subjects. A similar pattern was evident for erythrocyte GST activity in the GSTM1− subjects. No association was found among consumption of F&V and GSTM1/T1 genotypes and plasma vitamin C level. Different consumption of F&V had no impact on plasma T-AOC and vitamin C levels in the GSTM1−/GSTT1+ or GSTM1−/GSTT1− subjects. The erythrocyte GST activity was more sensitive to consumption of F&V in the individuals with the GSTM1−/GSTT1+ genotype. Association was found among GSTM1/T1 genotypes, antioxidant parameters and consumption of F&V. Large-scale and multiple ethnic studies are needed to further evaluate the relationship.
Collapse
|
43
|
Block G, Shaikh N, Jensen CD, Volberg V, Holland N. Serum vitamin C and other biomarkers differ by genotype of phase 2 enzyme genes GSTM1 and GSTT1. Am J Clin Nutr 2011; 94:929-37. [PMID: 21813807 PMCID: PMC3155929 DOI: 10.3945/ajcn.111.011460] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Glutathione S-transferases (GSTs) detoxify environmental chemicals and are involved in oxidative stress pathways. Deletion polymorphisms affect enzyme activities and have been associated with risk of disease. OBJECTIVE The objective was to clarify whether biomarkers of oxidation, antioxidation, inflammation, and nutritional factors differ by GST genotype in healthy adults. DESIGN Subjects (n = 383) consisted of nonsmokers and nonusers of antiinflammatory drugs and antioxidant vitamin supplements. Deletion polymorphisms of GSTM1 and GSTT1 were genotyped. F(2)-isoprostanes, malondialdehyde, C-reactive protein, serum vitamin C, carotenoids, tocopherols, and other nutritional factors were assessed. RESULTS The concentration of serum vitamin C was higher in persons with the inactive GSTM1-0 genotype (P = 0.006). This relation was unchanged after adjustment for age, sex, BMI, or dietary vitamin C. F(2)-isoprostanes and malondialdehyde were lower in the GSTM1-0 and GSTT1-0 groups, respectively, but significance was lost after control for serum vitamin C. The dual deletion, GSTM1-0/GSTT1-0 (n = 37), was associated with higher serum iron and total and LDL-cholesterol concentrations (all P < 0.01) and lower malondialdehyde concentrations, which persisted after adjustment for age, sex, BMI, and serum vitamin C. Carotenoids and α- and γ-tocopherols were not associated with either genotype. CONCLUSIONS Oxidative stress and inflammation biomarkers differ by GST genotype, but serum vitamin C appears to be the most consistent factor. Examination of other relevant genes may be needed to understand the concentration and function of ascorbic acid in the GST enzyme system. This trial is registered at clinicaltrials.gov as NCT00079963.
Collapse
Affiliation(s)
- Gladys Block
- School of Public Health, University of California at Berkeley, 94720-7360, USA.
| | | | | | | | | |
Collapse
|
44
|
NAD(P)H:Quinone Oxidoreductase 1 and its Potential Protective Role in Cardiovascular Diseases and Related Conditions. Cardiovasc Toxicol 2011; 12:39-45. [DOI: 10.1007/s12012-011-9136-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
45
|
Nørskov MS, Frikke-Schmidt R, Loft S, Sillesen H, Grande P, Nordestgaard BG, Tybjærg-Hansen A. Copy Number Variation in
Glutathione S-Transferases M1
and
T1
and Ischemic Vascular Disease. ACTA ACUST UNITED AC 2011; 4:418-28. [DOI: 10.1161/circgenetics.111.959809] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background—
Glutathione S-transferases (GSTs) M1 and T1 detoxify products of oxidative stress and may protect against atherosclerosis and ischemic vascular disease (IVD). We tested the hypothesis that copy number variation (CNV) in
GSTM1
and
GSTT1
genes, known to be associated with stepwise decreases in catalytic activity, predict risk of IVD.
Methods and Results—
We included 23 059 Danes from 2 general population studies and 2 case-control studies, of whom 4930 had ischemic heart disease (IHD) and 2086 had ischemic cerebrovascular disease. A real-time polymerase chain reaction method genotyped for the exact number of
GSTM1
and
GST
T1
gene copies. We also performed meta-analyses, including our own and former studies, totaling 13 196 IHD cases and 33 228 controls. CNV in
GSTM1
or
GSTT1
or genotype combinations were not associated with an increased risk of IHD, myocardial infarction, ischemic cerebrovascular disease, ischemic stroke, or any ischemic vascular event in studies individually or combined or in the meta-analyses. Furthermore, genotypes did not interact with smoking on risk of disease end points. Finally,
GST
genotypes did not associate with markers of inflammation and oxidation or interact with smoking on markers of inflammation in the general population. In contrast, we observed the well-established association between CNV in
GSTM1
and risk of bladder cancer.
Conclusions—
In studies including 6557 IVD cases and 16 502 controls and in meta-analyses of 13 196 cases and 33 228 controls, CNV in
GSTM1
and
GST
T1
genes did not associate with risk of IVD or with markers of inflammation. These observations were independent of smoking exposure.
Collapse
Affiliation(s)
- Marianne S. Nørskov
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Ruth Frikke-Schmidt
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Steffen Loft
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Henrik Sillesen
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Peer Grande
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Børge G. Nordestgaard
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| | - Anne Tybjærg-Hansen
- From the Departments of Clinical Biochemistry (M.S.N., R.F.-S., A.T.-H.), Vascular Surgery (H.S.), and Cardiology (P.G.), Rigshospitalet; Department of Environmental Health, Institute of Public Health (S.L.); Department of Clinical Biochemistry (B.G.N.) and The Copenhagen General Population Study (R.F.-S., B.G.N., A.T.-H.), Herlev Hospital; The Copenhagen City Heart Study (B.G.N., A.T.-H.), Bispebjerg Hospital; and Copenhagen University Hospitals and Faculty of Health Sciences (M.S.N., R.F.-S., S.L
| |
Collapse
|
46
|
Araujo ASR, Seibel FER, Oliveira UO, Fernandes T, Llesuy S, Kucharski L, Belló-Klein A. Thyroid hormone-induced haemoglobin changes and antioxidant enzymes response in erythrocytes. Cell Biochem Funct 2011; 29:408-13. [DOI: 10.1002/cbf.1765] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 03/22/2011] [Accepted: 04/12/2011] [Indexed: 11/06/2022]
Affiliation(s)
- A. S. R. Araujo
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| | - F. E. R. Seibel
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| | - U. O. Oliveira
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| | - T. Fernandes
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| | - S. Llesuy
- Catedra de Quimica General y Inorgánica. Facultad de Farmacia y Bioquimica; Universidad de Buenos Aires; Buenos Aires; Argentina
| | - L. Kucharski
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| | - A. Belló-Klein
- Cardiovascular Physiology Laboratory, Physiology Department; Basic Health Sciences Institute at Federal University of Rio Grande do Sul; Porto Alegre; Brazil
| |
Collapse
|
47
|
Potential risk modifications of GSTT1, GSTM1 and GSTP1 (glutathione-S-transferases) variants and their association to CAD in patients with type-2 diabetes. Biochem Biophys Res Commun 2011; 407:49-53. [PMID: 21352813 DOI: 10.1016/j.bbrc.2011.02.097] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 02/20/2011] [Indexed: 11/22/2022]
Abstract
BACKGROUND Type-2 diabetes mellitus (T2DM) is a major risk factor for coronary artery disease (CAD) resulting in high morbidity and mortality. Glutathione S-transferases (GSTM1, GSTT1 and GSTP1) are known for their broad range of detoxification and in the metabolism of xenobiotics. Recent studies revealed the relationship of GSTs variants with T2DM and CAD. In this case-control study we ascertained the association of GSTs variants in association with the development of CAD in patients with T2DM. METHODS From the Southern part of India, we enrolled 222 T2DM patients, 290 T2DM patients with CAD and 270 healthy controls matched for age, sex and origin. Serum lipid profiles were measured and DNA was extracted from the blood samples. Multiplex PCR for GSTM1/T1 (null polymorphism) and PCR-RFLP for GSTP1 (105 A>G), were performed for genotyping of study participants. Gene frequency and lipid profiles were statistically analyzed for disease association. RESULTS Regression analysis showed that, GSTM1-null genotype is associated with a 2-fold increase (OR=2.925; 95% CI=2.078-4.119; P<0.0001) and GSTT1-null genotype is associated with a 3-fold increase (OR=3.114; 95% CI=2.176-4.456; P<0.0001) to T2DM development. Ile/Val and Val/Val genotypes of GSTP1 also showed a significant risk for T2DM (OR=1.423, CI=1.041-1.946; P=0.027 and OR=1.829, CI=1.064-3.142; P=0.029). Increased odds ratio showed that GSTT1-null genotype had a moderately higher occurrence in T2DM-CAD patients (OR=1.918, 95% CI=1.144-3.214; P=0.014) than T2DM patients without CAD. The level of HDL has significantly decreased in GSTT1-present than in GSTT1-null genotype (43.50±4.10 vs. 45.20±3.90; P=0.004) when compared with control and T2DM patients. However, LDL level showed a significant increase in GSTT1-null than GSTT1-present genotype (108.70±16.90 vs. 102.20±12.60; P=0.005). 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). CONCLUSION Our work concludes that GSTM1, GSTT1 and GSTP1 variants might contribute to the development of T2DM and GSTT1 variant alone is involved in the development of T2DM associated CAD complications in the South Indian population.
Collapse
|
48
|
Hardwick RN, Fisher CD, Canet MJ, Lake AD, Cherrington NJ. Diversity in antioxidant response enzymes in progressive stages of human nonalcoholic fatty liver disease. Drug Metab Dispos 2010; 38:2293-301. [PMID: 20805291 PMCID: PMC2993454 DOI: 10.1124/dmd.110.035006] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 08/30/2010] [Indexed: 01/04/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), which occurs in approximately 17 to 40% of Americans, encompasses progressive stages of liver damage ranging from steatosis to nonalcoholic steatohepatitis (NASH). Inflammation and oxidative stress are known characteristics of NAFLD; however, the precise mechanisms occurring during disease progression remain unclear. The purpose of the current study was to determine whether the expression or function of enzymes involved in the antioxidant response, NAD(P)H:quinone oxidoreductase 1 (NQO1), glutathione transferase (GST), and glutamate cysteine ligase, are altered in the progression of human NAFLD. Human livers staged as normal, steatotic, NASH (fatty), and NASH (not fatty) were obtained from the Liver Tissue Cell Distribution System. NQO1 mRNA, protein, and activity tended to increase with disease progression. mRNA levels of the GST isoforms A1, A2, A4, M3, and P1 increased with NAFLD progression. Likewise, GST A and P protein increased with progression; however, GST M protein levels tended to decrease. Of interest, total GST activity toward the substrate 1-chloro-2,4-dinitrobenzene decreased with NAFLD progression. GSH synthesis does not seem to be significantly dysregulated in NAFLD progression; however, the GSH/oxidized glutathione redox ratio seemed to be reduced with disease severity, indicating the presence of oxidative stress and depletion of GSH throughout progression of NAFLD. Malondialdehyde concentrations were significantly increased with disease progression, further indicating the presence of oxidative stress. Nuclear immunohistochemical staining of nuclear factor E2-related factor 2 (Nrf2), an indicator of activation of the transcription factor, was evident in all stages of NAFLD. The current data suggest that Nrf2 activation occurs in response to disease progression followed by induction of specific Nrf2 targets, whereas functionality of specific antioxidant defense enzymes seems to be impaired as NAFLD progresses.
Collapse
Affiliation(s)
- Rhiannon N Hardwick
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona, USA
| | | | | | | | | |
Collapse
|
49
|
Wang J, Zou L, Huang S, Lu F, Lang X, Han L, Song Z, Xu Z. Genetic polymorphisms of glutathione S-transferase genes GSTM1, GSTT1 and risk of coronary heart disease. Mutagenesis 2010; 25:365-9. [PMID: 20354063 DOI: 10.1093/mutage/geq014] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
To clarify the role of glutathione S-transferases (GSTs; GSTM1 and GSTT1) status in susceptibility to coronary heart disease (CHD), a meta-analysis of published studies was performed. A total of 19 studies including 8020 cases and 11 501 controls were included in this meta-analysis. In a combined analysis, the relative risks for CHD of the GSTM1 null and GSTT1 null polymorphisms were 1.47 [95% confidence interval (CI): 1.08-2.01] and 1.26 (95% CI: 0.90-1.75), respectively. Three potential sources of heterogeneity including ethnicity, source of control and sample size of study were also assessed. However, no significant association was found in stratified analyses. By pooling data from eight studies (2909 cases and 3745 controls) that considered combinations of GSTT1 and GSTM1 genotypes, a statistically significant increased risk for CHD [odds ratio (OR = 2.38, 95% CI: 1.03-5.48)] was detected for individuals with combined deletion mutations in both genes compared with positive genotypes. Results from the meta-analysis of five studies on GSTs stratified according to smoking status showed an increased risk for individuals with null genotype (OR = 2.21, 95% CI: 1.24-3.92 for GSTM1 and OR = 3.29, 95% CI: 1.49-7.26 for GSTT1) versus non-null genotypes. This meta-analysis suggests that the GSTM1 null genotype may slightly increase the risk of CHD and that interaction between unfavourable GSTs genotypes may exist.
Collapse
Affiliation(s)
- Jun Wang
- Department of Cardiothoracic Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
50
|
da Rosa Araujo AS, Silva de Miranda MF, de Oliveira UO, Fernandes TÃ, Llesuy S, Rios Kucharski LC, Khaper N, Belló-Klein A. Increased resistance to hydrogen peroxide-induced cardiac contracture is associated with decreased myocardial oxidative stress in hypothyroid rats. Cell Biochem Funct 2010; 28:38-44. [DOI: 10.1002/cbf.1616] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|