1
|
Appell ML, Hindorf U, Almer S, Haglund S. Response to azathioprine treatment in autoimmune hepatitis is dependent on glutathione transferase genotypes. Dig Liver Dis 2025; 57:885-892. [PMID: 39863504 DOI: 10.1016/j.dld.2024.12.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/23/2024] [Accepted: 12/24/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND Azathioprine (AZA) is part of the standard treatment for autoimmune hepatitis (AIH). The first step in the complex bioconversion of AZA to active metabolites is mediated by glutathione transferases (GSTs). AIMS Elucidate the association between GSTM1 and GSTT1 copy number variation (CNV), genetic variation in GSTA2, GSTP1, and inosine-triphosphate-pyrophosphatase, and the response to AZA in AIH. METHODS Genotyping was performed in AIH patients (n = 131) on AZA, and in a Swedish background population (n = 283). Thiopurine metabolites in blood erythrocytes were determined by high performance liquid chromatography. RESULTS GSTM1 and GSTT1 CNV were associated with treatment response to AZA. Gene deletion of GSTM1-but not of GSTT1-was associated with the liver transaminase levels. None of the studied genetic variants were associated with the thiopurine metabolite concentrations, suggesting non-enzymatic mechanisms of GSTM1 and GSTT1 in the context of AZA efficacy in AIH. The prevalence of GSTM1 and GSTT1 CNV genotypes was similar in AIH and in the background population. CONCLUSION This study shows the effects of GSTM1 and GSTT1 CNV on AZA efficacy in AIH, not previously described. It also elaborates on the impact of the definition of treatment response, on the importance of the various GSTs studied. Furthermore, the GSTM1 and GSTT1 CNV frequencies previously reported in European populations were confirmed.
Collapse
Affiliation(s)
- Malin Lindqvist Appell
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| | - Ulf Hindorf
- Department of Gastroenterology and Nutrition, University Hospital Lund, Lund, Sweden.
| | - Sven Almer
- Centre for Digestive Health, Department of Gastroenterology, Dermatology and Rheumatology, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine, Solna, Karolinska Institutet, Stockholm, Sweden.
| | - Sofie Haglund
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden; Department of Laboratory Medicine, Region Jönköping County, Jönköping, Sweden.
| |
Collapse
|
2
|
Zheng Y, Tang M, Deng Z, Cai P. Genetic polymorphisms and platinum-induced hematological toxicity: a systematic review. Front Pharmacol 2024; 15:1445328. [PMID: 39234108 PMCID: PMC11371761 DOI: 10.3389/fphar.2024.1445328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Background Platinum-based chemotherapy bring severe hematological toxicity that can lead to dose reduction or discontinuation of therapy. Genetic variations have been reported to influence the risk and extent of hematological toxicity; however, the results are controversial and a comprehensive overview is lacking. This systematic review aimed to identify genetic biomarkers of platinum-induced hematological toxicity. Method Pubmed, Embase and Web of science database were systematically reviewed for studies that evaluated the association of genetic variants and platinum-related hematological toxicity in tumor patients with no prior history of chemotherapy or radiation, published from inception to the 28th of January 2022. The studies should have specific toxicity scoring system as well as defined toxicity end-point. The quality of reporting was assessed using the Strengthening the Reporting of Genetic Association Studies (STREGA) checklist. Results were summarized using narrative synthesis. Results 83 studies were eligible with over 682 single-nucleotide polymorphisms across 110 genes. The results are inconsistent and diverse with methodological issues including insufficient sample size, population stratification, various treatment schedule and toxicity end-point, and inappropriate statistics. 11 SNPs from 10 genes (ABCB1 rs1128503, GSTP1 rs1695, GSTM1 gene deletion, ERCC1 rs11615, ERCC1 rs3212986, ERCC2 rs238406, XPC rs2228001, XPCC1 rs25487, MTHFR rs1801133, MDM2 rs2279744, TP53 rs1042522) had consistent results in more than two independent populations. Among them, GSTP1 rs1695, ERCC1 rs11615, ERCC1 rs3212986, and XRCC1 rs25487 present the most promising results. Conclusion Even though the results are inconsistent and several methodological concerns exist, this systematic review identified several genetic variations that deserve validation in well-defined studies with larger sample size and robust methodology. Systematic Review Registration https://www.crd.york.ac.uk/, identifier CRD42021234164.
Collapse
Affiliation(s)
- Yi Zheng
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| | - Mimi Tang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng Deng
- Hunan Institute for Tuberculosis Control and Hunan Chest Hospital, Changsha, China
- Hunan Chest Hospital, Changsha, China
| | - Pei Cai
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, China
| |
Collapse
|
3
|
Sehgal P, Chaturvedi P. Chromatin and Cancer: Implications of Disrupted Chromatin Organization in Tumorigenesis and Its Diversification. Cancers (Basel) 2023; 15:cancers15020466. [PMID: 36672415 PMCID: PMC9856863 DOI: 10.3390/cancers15020466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
A hallmark of cancers is uncontrolled cell proliferation, frequently associated with an underlying imbalance in gene expression. This transcriptional dysregulation observed in cancers is multifaceted and involves chromosomal rearrangements, chimeric transcription factors, or altered epigenetic marks. Traditionally, chromatin dysregulation in cancers has been considered a downstream effect of driver mutations. However, here we present a broader perspective on the alteration of chromatin organization in the establishment, diversification, and therapeutic resistance of cancers. We hypothesize that the chromatin organization controls the accessibility of the transcriptional machinery to regulate gene expression in cancerous cells and preserves the structural integrity of the nucleus by regulating nuclear volume. Disruption of this large-scale chromatin in proliferating cancerous cells in conventional chemotherapies induces DNA damage and provides a positive feedback loop for chromatin rearrangements and tumor diversification. Consequently, the surviving cells from these chemotherapies become tolerant to higher doses of the therapeutic reagents, which are significantly toxic to normal cells. Furthermore, the disorganization of chromatin induced by these therapies accentuates nuclear fragility, thereby increasing the invasive potential of these tumors. Therefore, we believe that understanding the changes in chromatin organization in cancerous cells is expected to deliver more effective pharmacological interventions with minimal effects on non-cancerous cells.
Collapse
|
4
|
Levy R, Le TH. Role of GSTM1 in Hypertension, CKD, and Related Diseases across the Life Span. KIDNEY360 2022; 3:2153-2163. [PMID: 36591365 PMCID: PMC9802555 DOI: 10.34067/kid.0004552022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 10/17/2022] [Indexed: 12/31/2022]
Abstract
Over 20 years after the introduction of angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, CKD remains a major public health burden with limited therapeutic options to halt or slow kidney disease progression at all ages. The consensus is that oxidative stress contributes to CKD development and progression. Yet, to date, there is no clear evidence that broad use of antioxidant therapy provides a beneficial effect in CKD. Understanding the specific pathophysiologic mechanisms in those who are genetically most susceptible to oxidative stress is a crucial step to inform therapy in an individualized medicine approach, considering differing exposures and risks across the life span. Glutathione-S-transferase μ 1 (GSTM1) is a phase 2 enzyme involved in inactivation of reactive oxygen species and metabolism of xenobiotics. In particular, those with the highly prevalent GSTM1 null genotype (GSTM1[0/0]) may be more susceptible to kidney disease progression, due to impaired capacity to handle the increased oxidative stress burden in disease states, and might specifically benefit from therapy that targets the redox imbalance mediated by loss of the GSTM1 enzyme. In this review, we will discuss the studies implicating the role of GSTM1 deficiency in kidney and related diseases from experimental rodent models to humans, from the prenatal period through senescence, and the potential underlying mechanism.
Collapse
Affiliation(s)
- Rebecca Levy
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| | - Thu H. Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, Rochester, New York
| |
Collapse
|
5
|
Moody L, Xu GB, Pan YX, Chen H. Genome-wide cross-cancer analysis illustrates the critical role of bimodal miRNA in patient survival and drug responses to PI3K inhibitors. PLoS Comput Biol 2022; 18:e1010109. [PMID: 35639779 PMCID: PMC9187341 DOI: 10.1371/journal.pcbi.1010109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/10/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022] Open
Abstract
Heterogeneity of cancer means many tumorigenic genes are only aberrantly expressed in a subset of patients and thus follow a bimodal distribution, having two modes of expression within a single population. Traditional statistical techniques that compare sample means between cancer patients and healthy controls fail to detect bimodally expressed genes. We utilize a mixture modeling approach to identify bimodal microRNA (miRNA) across cancers, find consistent sources of heterogeneity, and identify potential oncogenic miRNA that may be used to guide personalized therapies. Pathway analysis was conducted using target genes of the bimodal miRNA to identify potential functional implications in cancer. In vivo overexpression experiments were conducted to elucidate the clinical importance of bimodal miRNA in chemotherapy treatments. In nine types of cancer, tumors consistently displayed greater bimodality than normal tissue. Specifically, in liver and lung cancers, high expression of miR-105 and miR-767 was indicative of poor prognosis. Functional pathway analysis identified target genes of miR-105 and miR-767 enriched in the phosphoinositide-3-kinase (PI3K) pathway, and analysis of over 200 cancer drugs in vitro showed that drugs targeting the same pathway had greater efficacy in cell lines with high miR-105 and miR-767 levels. Overexpression of the two miRNA facilitated response to PI3K inhibitor treatment. We demonstrate that while cancer is marked by considerable genetic heterogeneity, there is between-cancer concordance regarding the particular miRNA that are more variable. Bimodal miRNA are ideal biomarkers that can be used to stratify patients for prognosis and drug response in certain types of cancer. Bimodal genes can be defined as those having two modes of expression within the same population. A variety of statistical methodologies have been employed to assess bimodal gene expression, but current methods and their applications have been limited. Given the advances in next-generation sequencing as well as the extensive regulatory role of miRNA, assessing bimodality in miRNA-seq data can greatly broaden our understanding of factors underlying tumor progression. The goal of the current study was to utilize a novel mixture modeling approach to identify bimodal miRNA and then demonstrate their importance in cancer by evaluating their ability to predict overall survival and drug response. Our results showed that high levels of bimodal miRNA expression was characteristic of cancer. Additionally, several bimodal miRNA were common to multiple cancer types, suggesting that certain miRNA consistently account for tumor heterogeneity and may be involved in general oncogenic processes. Our study points to the potential of bimodal miRNA to facilitate precise prognostic evaluation and effective treatment strategies.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Guanying Bianca Xu
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- * E-mail:
| |
Collapse
|
6
|
Hemlata, Singh J, Bhardwaj A, Kumar A, Singh G, Priya K, Giri SK. Comparative frequency distribution of glutathione S-transferase mu (GSTM1) and theta (GSTT1) allelic forms in Himachal Pradesh population. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00298-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Glutathione S-transferases (GSTs) are a class of important Phase II detoxification enzymes that catalyze the conjugation of glutathione and xenobiotic compounds (environmental carcinogens, pollutants and drugs) to protect against oxidative stress. GSTT1 and GSTM1 genetic polymorphisms have been extensively studied, and null genotypes or homozygous deletions have been reported in various populations. Previous studies have suggested that those who are homozygous null at the GSTM1 or GSTT1 loci are more susceptible and have a higher risk of cancers linked to environmental pollutants and drug-induced toxicity. Our study focused on GSTM1 and GSTT1 null allele frequency in the Doon population of Himachal Pradesh (India) with a comparison across other Inter and Intra-Indian ethnic groups to predict variation in the possible susceptible status.
Material and methods
Genomic DNA samples were extracted from 297 healthy unrelated individuals by a ReliaPrep™ Blood gDNA Miniprep kit (Promega, USA), and genotyped for allelic variation in GSTM1 and GSTT1 genotypes by multiplex polymerase chain reaction. Fisher's exact test was applied using SPSS.20 to analyze the genotypic distribution of GSTM1 and GSTT1 null alleles in male and female of Doon region (Solan) Himachal Pradesh.
Results
In our study, the frequency distribution of the homozygous null genotypes of GSTM1, GSTT1 individually as well as combined was found as 33.3%, 32% and 9%, respectively. Upon gender-wise comparison, a non-significant distribution (p > 0.05) for null genotypes of GSTM1 (32.8% and 35.4%, OR-0.77, 95% CI 0.42–1.41), GSTT1 (33.2% and 27.7%, OR-1.12, 95% CI 0.63–2.0) individually and combined GSTM1 and GSTT1 (10.8% and 3.7%, OR-0.31, 95% CI 0.07–1.42) were observed in studied population.
Conclusions
In our studied population, the frequency of GSTM1 null genotypes was found deviated from Inter- and Intra-Indian ethnic groups. However, the frequency of homozygous null type of GSTT1 was not significantly different, when compared to previous Indian studies, comparison with global ethnic groups showed deviation. Thus, our study has highlighted possible susceptibility risk to various xenobiotics in the Doon population of Himachal Pradesh, India.
Collapse
|
7
|
Effect of GSTA1 Variants on Busulfan-Based Conditioning Regimen Prior to Allogenic Hematopoietic Stem-Cell Transplantation in Pediatric Asians. Pharmaceutics 2022; 14:pharmaceutics14020401. [PMID: 35214132 PMCID: PMC8880478 DOI: 10.3390/pharmaceutics14020401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Busulfan is widely used as a chemotherapy treatment before hematopoietic stem-cell transplantation (HSCT). However, the response of busulfan is highly variable and unpredictable, whereby the pharmacogenetic interference of glutathione S-transferase (GST) has strong evidence in Caucasians and some adult Asians but not in pediatric Asian patients. This study was aimed at investigating the associations of GST genetic polymorphisms with variations in the pharmacokinetic (PK) properties of busulfan in pediatric Asian patients. This retrospective cohort study recruited 92 pediatric patients. The polymorphism of GSTA1 was genotyped by Sanger sequencing, and GSTM1 and GSTP1 were genotyped by real-time PCR. Drug concentration and PK estimation were identified using an LC-MS/MS method and a noncompartmental model. Statistical analysis was performed by R software. Out of 92 patients, 48 (53%) were males, the mean age was 8.4 ± 5.12 years old, and the average weight was 26.52 ± 14.75 kg. The allele frequencies of GSTA1*B and of GSTM1 and GSTP1* deletions were 16.9%, 68.5%, and 21.2%, respectively. Patients with GSTA1*B had a statistically significant impact on the PK of busulfan, whereas those with GSTM1 and GSTP1 did not (p > 0.05). The carriers of GSTA1*B showed a significant difference compared to noncarriers in terms of t1/2 (for first dose: 161.9 vs. 134.3 min, p = 0.0016; for second dose: 156.1 vs. 129.8, p = 0.012), CL (88.74 vs. 124.23 mL/min, p = 0.0089), Cmax (4232.6 vs. 3675.5 ng/mL, p = 0.0021), and AUC (5310.6 vs. 4177.1 µM/min, p = 0.00033). The augmentation of AUC was around 27.1% in patients carrying the GSTA1*B variant. The GSTA1 polymorphism was significantly associated with variations of the pharmacokinetic properties of busulfan treatment in pediatric Asian patients.
Collapse
|
8
|
García-González I, Pérez-Mendoza G, Solís-Cárdenas A, Flores-Ocampo J, Herrera-Sánchez LF, Mendoza-Alcocer R, González-Herrera L. Genetic variants of PON1, GSTM1, GSTT1, and locus 9p21.3, and the risk for premature coronary artery disease in Yucatan, Mexico. Am J Hum Biol 2021; 34:e23701. [PMID: 34766662 DOI: 10.1002/ajhb.23701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/22/2021] [Accepted: 11/01/2021] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVE Genetic variants of PON1, rs70587, rs662, rs854560, GSTM1, and GSTT1 and two single nucleotide polymorphisms (SNP) at 9p21.3 locus, rs1333049, and rs2383207; were evaluated in association with the risk for premature coronary artery disease (CAD) in a population of Yucatan, Mexico. These genes are involved in the inactivation of pro-oxidants and pro-inflammatory mediators, lipid and xenobiotic metabolism, detoxification of reactive oxygen species, and regulation of cellular proliferation playing key roles in the pathogenesis of atherosclerosis. METHODS We conducted a matched case-control study with 98 CAD cases and 101 healthy controls. Genotyping of PON1 and 9p21.2 SNP was performed by real time-PCR and for GSTM1 and GSTT1 with multiplex-PCR. Odds ratios (OR) were calculated to estimate association and generalized multifactor dimensionality reduction (GMDR) algorithm to identify gene-gene and gene-environment interactions. RESULTS The distribution of all allele/genotype frequencies in controls was within Hardy-Weinberg expectations (p > .05) except for GSTM1. The allele/genotype frequencies of the GSTT1 null were significantly higher in CAD cases than in controls, suggesting association with higher risk for developing CAD. The other SNPs did not show any significant independent association with premature CAD. GMDR revealed a significant interaction between GSTT1 and LL55 genotype. Likewise, the body mass index (BMI) and smoking also showed an interaction with GSTT1. CONCLUSION The GSTT1 null allele/genotype is associated with an increased risk of developing premature CAD, the effect of which is not modified by cardiovascular risk factors in the population of Yucatan.
Collapse
Affiliation(s)
- Igrid García-González
- Laboratorio de Genética, Centro de Investigaciones Regionales 'Dr. Hideyo Noguchi', Universidad Autónoma de Yucatán (UADY), Mérida, Yucatán, Mexico
| | - Gerardo Pérez-Mendoza
- Laboratorio de Genética, Centro de Investigaciones Regionales 'Dr. Hideyo Noguchi', Universidad Autónoma de Yucatán (UADY), Mérida, Yucatán, Mexico
| | | | - Jorge Flores-Ocampo
- Servicio de Cardiología, Hospital Regional del ISSSTE, Mérida, Yucatán, Mexico
| | | | - Renan Mendoza-Alcocer
- Centro Estatal de la transfusión sanguínea, Servicios de Salud de Yucatán, Mérida, Yucatán, Mexico
| | - Lizbeth González-Herrera
- Laboratorio de Genética, Centro de Investigaciones Regionales 'Dr. Hideyo Noguchi', Universidad Autónoma de Yucatán (UADY), Mérida, Yucatán, Mexico
| |
Collapse
|
9
|
Alvarado AT, Muñoz AM, Bartra MS, Valderrama-Wong M, González D, Quiñones LA, Varela N, Bendezú MR, García JA, Loja-Herrera B. Frequency of CYP1A1*2A polymorphisms and deletion of the GSMT1 gene in a Peruvian mestizo population. PHARMACIA 2021. [DOI: 10.3897/pharmacia.68.e71621] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The polymorphic variants of CYP1A1 and the deletion of GSTM1 are present in the Peruvian mestizo population. Wild type and mutated genotypes (WT/*2A and *2A/ *2A) were identified, whose allele frequencies are 0.31 (T allele) and 0.69 (C allele), respectively; 53% with wild type GSTM1 (+) and 47% with null GSTM1. The frequency in Iquiteño emigrants was 0.72 CYP1A1*2A and 25% GSTM1 (-); from Lima 0.67 CYP1A1*2A and 33% of GSTM1 (-). The Hardy-Weinberg equilibrium test for the studied population showed that both frequencies are out of balance, p > .05.
The presence of the risk allele of the CYP1A1*2A polymorphism and the deletion in the GSTM1 gene are high, which could be indicative of a phase I and II metabolic imbalance in this group of Peruvian populations, with potential risks of activating agents procarcinogens thus affecting the incidence of tumor pathologies with an environmental component.
Collapse
|
10
|
Le TH. GSTM1 Gene, Diet, and Kidney Disease: Implication for Precision Medicine?: Recent Advances in Hypertension. Hypertension 2021; 78:936-945. [PMID: 34455814 DOI: 10.1161/hypertensionaha.121.16510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the United States, the prevalence of chronic kidney disease in adults is ≈14%. The mainstay of therapy for chronic kidney disease is angiotensin-converting enzyme inhibitors or angiotensin receptor blockers, but many patients with chronic kidney disease still progress to end-stage kidney disease. Increased oxidative stress is a major molecular underpinning of chronic kidney disease progression. In humans, a common deletion variant of the glutathione-S-transferase μ-1 (GSTM1) gene, the GSTM1 null allele (GSTM1(0)), results in decreased GSTM1 enzymatic activity and is associated with higher levels of oxidative stress. GSTM1 belongs to the superfamily of GSTs that are phase II antioxidant enzymes and are regulated by Nrf2 (nuclear factor erythroid 2-related factor 2). Cruciferous vegetables in general, and broccoli in particular, are rich in glucoraphanin, a precursor of sulforaphane that has been shown to have protective effects against oxidative damage through the activation of Nrf2. This review will highlight recent human and animal studies implicating the role of GSTM1 deficiency in hypertension and kidney disease, and its impact on the effects of cruciferous vegetables on kidney injury and disease progression, illustrating the significance of gene and environment interaction and a potential for targeted precision medicine in the treatment of kidney disease.
Collapse
Affiliation(s)
- Thu H Le
- Division of Nephrology, Department of Medicine, University of Rochester Medical Center, NY
| |
Collapse
|
11
|
van de Wetering C, Elko E, Berg M, Schiffers CHJ, Stylianidis V, van den Berge M, Nawijn MC, Wouters EFM, Janssen-Heininger YMW, Reynaert NL. Glutathione S-transferases and their implications in the lung diseases asthma and chronic obstructive pulmonary disease: Early life susceptibility? Redox Biol 2021; 43:101995. [PMID: 33979767 PMCID: PMC8131726 DOI: 10.1016/j.redox.2021.101995] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 01/01/2023] Open
Abstract
Our lungs are exposed daily to airborne pollutants, particulate matter, pathogens as well as lung allergens and irritants. Exposure to these substances can lead to inflammatory responses and may induce endogenous oxidant production, which can cause chronic inflammation, tissue damage and remodeling. Notably, the development of asthma and Chronic Obstructive Pulmonary Disease (COPD) is linked to the aforementioned irritants. Some inhaled foreign chemical compounds are rapidly absorbed and processed by phase I and II enzyme systems critical in the detoxification of xenobiotics including the glutathione-conjugating enzymes Glutathione S-transferases (GSTs). GSTs, and in particular genetic variants of GSTs that alter their activities, have been found to be implicated in the susceptibility to and progression of these lung diseases. Beyond their roles in phase II metabolism, evidence suggests that GSTs are also important mediators of normal lung growth. Therefore, the contribution of GSTs to the development of lung diseases in adults may already start in utero, and continues through infancy, childhood, and adult life. GSTs are also known to scavenge oxidants and affect signaling pathways by protein-protein interaction. Moreover, GSTs regulate reversible oxidative post-translational modifications of proteins, known as protein S-glutathionylation. Therefore, GSTs display an array of functions that impact the pathogenesis of asthma and COPD. In this review we will provide an overview of the specific functions of each class of mammalian cytosolic GSTs. This is followed by a comprehensive analysis of their expression profiles in the lung in healthy subjects, as well as alterations that have been described in (epithelial cells of) asthmatics and COPD patients. Particular emphasis is placed on the emerging evidence of the regulatory properties of GSTs beyond detoxification and their contribution to (un)healthy lungs throughout life. By providing a more thorough understanding, tailored therapeutic strategies can be designed to affect specific functions of particular GSTs.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Marijn Berg
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Caspar H J Schiffers
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Vasili Stylianidis
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Maarten van den Berge
- Pulmonology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Martijn C Nawijn
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
12
|
Mawick A, Pfeiffer H, Vennemann M. Sudden infant death syndrome: deletions of glutathione-S-transferase genes M1 and T1 and tobacco smoke exposure. Int J Legal Med 2021; 135:1375-1383. [PMID: 33934228 PMCID: PMC8206056 DOI: 10.1007/s00414-021-02556-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/26/2021] [Indexed: 11/30/2022]
Abstract
In developed countries, sudden infant death syndrome (SIDS) is the leading cause of death in infants in their first year of life. The risk of SIDS is increased if parents smoked during pregnancy and in presence of the child. Glutathione S-transferases (GSTs) catalyse the conjugation of glutathione with electrophilic compounds and toxins, making them less reactive and easier to excrete. As a gene dose effect was observed for GSTM1 and GSTT1, the aim of this study was to investigate whether there is a connection between homozygous or heterozygous gene deletions of GSTM1 or GSTT1 and the occurrence of SIDS. We found that heterozygous deletion of GSTM1 occurred significantly more frequently in the SIDS case group compared to the control group. A homozygous deletion of GSMT1 was slightly more frequently in the control group. A homozygous gene deletion of GSTT1 showed no significant difference between the SIDS group and the control group. We also found that in the SIDS group, the number of victims that were exposed to cigarette smoke was significantly higher than the number of victims without cigarette smoke exposure and that the mean lifetime of children whose mothers smoked was shorter in comparison with non-smoking mothers. In SIDS cases with homozygous gene deletions of GSTM1, the median life span of children with tobacco smoke exposure was 60 days shorter than without smoke exposure. In conclusion, the absence of these two genes is not the only trigger for SIDS but could be a critical aspect of SIDS aetiology, particularly in SIDS cases with smoking parents.
Collapse
Affiliation(s)
- Anthea Mawick
- Institute of Legal Medicine, University Hospital of Münster, Röntgenstr. 23, 48149, Münster, Germany
| | - Heidi Pfeiffer
- Institute of Legal Medicine, University Hospital of Münster, Röntgenstr. 23, 48149, Münster, Germany
| | - Marielle Vennemann
- Institute of Legal Medicine, University Hospital of Münster, Röntgenstr. 23, 48149, Münster, Germany.
| |
Collapse
|
13
|
Almeida M, Soares M, Fonseca-Moutinho J, Ramalhinho AC, Breitenfeld L. Influence of Estrogenic Metabolic Pathway Genes Polymorphisms on Postmenopausal Breast Cancer Risk. Pharmaceuticals (Basel) 2021; 14:ph14020094. [PMID: 33513690 PMCID: PMC7910923 DOI: 10.3390/ph14020094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 01/23/2021] [Indexed: 12/21/2022] Open
Abstract
Estrogen metabolism plays an important role in tumor initiation and development. Lifetime exposure to high estrogens levels and deregulation of enzymes involved in estrogen biosynthetic and metabolic pathway are considered risk factors for breast cancer. The present study aimed to evaluate the impact of mutations acquisition during the lifetime in low penetrance genes that codify enzymes responsible for estrogen detoxification. Genotype analysis of GSTM1 and GSTT1 null polymorphisms, CYP1B1 Val432Leu and MTHFR C677T polymorphisms was performed in 157 samples of women with hormone-dependent breast cancer and correlated with the age at diagnosis. The majority of patients with GSTT1 null genotype and with both GSTM1 and GSTT1 null genotypes were 50 years old or more at the diagnosis (p-value = 0.021 and 0.018, respectively). Older women with GSTM1 null genotype were also carriers of the CYP1B1Val allele (p-value = 0.012). As well, GSTT1 null and CYP1B1Val genotypes were correlated with diagnosis at later ages (p-value = 0.022). Similar results were found associating MTHFR C677T and GSTT1 null polymorphism (p-value = 0.034). Our results suggest that estrogen metabolic pathway polymorphisms constitute a factor to be considered simultaneously with models for breast cancer risk assessment.
Collapse
Affiliation(s)
- Micaela Almeida
- Health Sciences Research Centre (CICS), Faculty of Health Sciences, University of Beira Interior (UBI), Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (M.A.); (M.S.); (J.F.-M.); (A.C.R.)
| | - Mafalda Soares
- Health Sciences Research Centre (CICS), Faculty of Health Sciences, University of Beira Interior (UBI), Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (M.A.); (M.S.); (J.F.-M.); (A.C.R.)
| | - José Fonseca-Moutinho
- Health Sciences Research Centre (CICS), Faculty of Health Sciences, University of Beira Interior (UBI), Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (M.A.); (M.S.); (J.F.-M.); (A.C.R.)
- Academic Hospital of Cova da Beira (CHUCB), Quinta do Alvito, 6200-251 Covilhã, Portugal
| | - Ana Cristina Ramalhinho
- Health Sciences Research Centre (CICS), Faculty of Health Sciences, University of Beira Interior (UBI), Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (M.A.); (M.S.); (J.F.-M.); (A.C.R.)
- Academic Hospital of Cova da Beira (CHUCB), Quinta do Alvito, 6200-251 Covilhã, Portugal
| | - Luiza Breitenfeld
- Health Sciences Research Centre (CICS), Faculty of Health Sciences, University of Beira Interior (UBI), Avenida Infante D. Henrique, 6200-506 Covilhã, Portugal; (M.A.); (M.S.); (J.F.-M.); (A.C.R.)
- Correspondence: ; Tel.: +351-2753-290-51
| |
Collapse
|
14
|
Ferracini AC, Lopes-Aguiar L, Lourenço GJ, Yoshida A, Lima CSP, Sarian LO, Derchain S, Kroetz DL, Mazzola PG. GSTP1 and ABCB1 Polymorphisms Predicting Toxicities and Clinical Management on Carboplatin and Paclitaxel-Based Chemotherapy in Ovarian Cancer. Clin Transl Sci 2020; 14:720-728. [PMID: 33326171 PMCID: PMC7993324 DOI: 10.1111/cts.12937] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Variation in drug disposition genes might contribute to susceptibility to toxicities and interindividual differences in clinical management on chemotherapy for epithelial ovarian cancer (EOC). This study was designed to explore the association of GST and ABCB1 genetic variation with hematologic and neurologic toxicity, changes in chemotherapy, and disease prognosis in Brazilian women with EOC. A total of 112 women with a confirmed histological diagnosis of EOC treated with carboplatin/paclitaxel were enrolled (2014–2019). The samples were analyzed by multiplex polymerase chain reaction (PCR) for the deletion of GSTM1 and GSTT1 genes. GSTP1 (c.313A>G/rs1695) and ABCB1 (c.1236C>T/rs1128503; c.3435C>T/rs1045642; c.2677G>T>A/rs2032582) single nucleotide polymorphisms (SNPs) were detected by real‐time PCR. Subjects with the GSTP1 c.313A>G had reduced risk of anemia (odds ratio (OR): 0.17, 95% confidence interval (CI): 0.04–0.69, P = 0.01, dominant model) and for thrombocytopenia (OR: 0.27, 95% CI: 0.12–0.64, P < 0.01; OR 0.18, 95% CI 0.03–0.85, P = 0.03, either dominant or recessive model), respectively. The GSTP1 c.313A>G AG genotype was associated with a lower risk of dose delay (OR: 0.35, 95% CI: 0.13–0.90, P = 0.03). The ABCB1 c.1236C>T was associated with increased risk of thrombocytopenia (OR: 0.15, 95% CI: 0.03–0.82, P = 0.03), whereas ABCB1 c.3435C>T had increased risk of grade 2 and 3 neurotoxicity (OR: 3.61, 95% CI: 1.08–121.01, P = 0.03) in recessive model (CC + CT vs. TT). This study suggests that GSTP1 c.313A>G, ABCB1 c.1236C>T, and c.3435C>T SNP detection is a potential predictor of hematological toxicity and neurotoxicity and could help predict the clinical management of women with EOC.
Collapse
Affiliation(s)
- Amanda Canato Ferracini
- Postgraduate Program in Medical Sciences, Faculty of Medical Sciences, University of Campinas, Campinas, Brazil.,Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | - Leisa Lopes-Aguiar
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Gustavo Jacob Lourenço
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Adriana Yoshida
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Carmen Silva Passos Lima
- Laboratory of Cancer Genetics, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Luis Otávio Sarian
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Sophie Derchain
- Department of Obstetrics and Gynecology, Faculty of Medical Sciences, State University of Campinas, Campinas, Brazil
| | - Deanna L Kroetz
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
15
|
Sljivancanin Jakovljevic T, Jacimovic J, Nikolic N, Milasin J. Lack of association between glutathione S-transferase M1 and T1 gene polymorphisms and susceptibility to preeclampsia: An updated systematic review and meta-analysis. Am J Reprod Immunol 2020; 84:e13303. [PMID: 32658338 DOI: 10.1111/aji.13303] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/05/2020] [Accepted: 07/09/2020] [Indexed: 01/23/2023] Open
Abstract
Insufficient response to oxidative stress in placenta is proposed as a contributing factor for preeclampsia (PE) development. Glutathione S-transferases (GST) have significant role in detoxification processes. Conflicting results were published by several research groups regarding GST T1 and GST M1 deletion polymorphism as risk factors for PE. The aim of the present meta-analysis was to get a better understanding of the impact of these polymorphisms in preeclampsia development. To identify relevant case-control studies, the author team searched Clarivate Analytics Web of Science, Scopus, PubMed, Cochrane Central Register of Controlled Trials, China National Knowledge Infrastructure, major subject journals, and gray literature. Pooled odds ratios and 95% confidence intervals for GST M1 and GST T1 deletion polymorphism and preeclampsia were derived from random effects models. This meta-analysis included 10 eligible studies. The pooled analyses showed no association between GST M1/GST T1 deletion polymorphisms and susceptibility to PE. Even though high heterogeneity was founded among results for GST M1 and double null genotypes, Egger's and Begg's tests (0.17 and 0.18, respectively) revealed no statistical evidence of publication bias among included studies. The present updated systematic review and meta-analysis found no association between GST M1 and GST T1 deletion polymorphism and PE risk.
Collapse
Affiliation(s)
| | - Jelena Jacimovic
- Central Library, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Nadja Nikolic
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - Jelena Milasin
- Department of Human Genetics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
16
|
Singh P, Wang X, Hageman L, Chen Y, Magdy T, Landier W, Ginsberg JP, Neglia JP, Sklar CA, Castellino SM, Dreyer ZE, Hudson MM, Robison LL, Blanco JG, Relling MV, Burridge P, Bhatia S. Association of GSTM1 null variant with anthracycline-related cardiomyopathy after childhood cancer-A Children's Oncology Group ALTE03N1 report. Cancer 2020; 126:4051-4058. [PMID: 32413235 PMCID: PMC7423633 DOI: 10.1002/cncr.32948] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/07/2019] [Accepted: 01/03/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Anthracycline-related cardiomyopathy is a leading cause of late morbidity in childhood cancer survivors. Glutathione S-transferases (GSTs) are a class of phase II detoxification enzymes that facilitate the elimination of anthracyclines. As free-radical scavengers, GSTs could play a role in oxidative damage-induced cardiomyopathy. Associations between the GSTμ1 (GSTM1) null genotype and iron-overload-related cardiomyopathy have been reported in patients with thalassemia. METHODS The authors sought to identify an association between the GSTM1 null genotype and anthracycline-related cardiomyopathy in childhood cancer survivors and to corroborate the association by examining GSTM1 gene expression in peripheral blood and human-induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) from survivors with and without cardiomyopathy. GSTM1 gene deletion was examined by polymerase chain reaction in 75 survivors who had clinically validated cardiomyopathy (cases) and in 92 matched survivors without cardiomyopathy (controls). Conditional logistic regression analysis adjusting for sex, age at cancer diagnosis, chest radiation, and anthracycline dose was used to assess the association between genotype and cardiomyopathy. Proprietary bead array technology and quantitative real-time polymerase chain reaction were used to measure GSTM1 expression levels in samples from 20 cases and 20 matched controls. hiPSC-CMs from childhood cancer survivors (3 with cardiomyopathy, 3 without cardiomyopathy) also were examined for GSTM1 gene expression levels. RESULTS A significant association was observed between the risk of cardiomyopathy and the GSTM1 null genotype (odds ratio, 2.7; 95% CI, 1.3-5.9; P = .007). There was significant downregulation of GSTM1 expression in cases compared with controls (average relative expression, 0.67 ± 0.57 vs 1.33 ± 1.33, respectively; P = .049). hiPSC-CMs from patients who had cardiomyopathy revealed reduced GSTM1 expression (P = .007). CONCLUSIONS The current findings could facilitate the identification of childhood cancer survivors who are at risk for anthracycline-related cardiomyopathy.
Collapse
Affiliation(s)
- Purnima Singh
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton, Texas
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jill P. Ginsberg
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Joseph P. Neglia
- Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota
| | - Charles A. Sklar
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sharon M. Castellino
- Department of Pediatrics, Emory University and Children’s Healthcare of Atlanta, Atlanta, Georgia
| | - Zoann E. Dreyer
- Department of Pediatrics, Texas Children’s Cancer Center, Houston, Texas
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Leslie L. Robison
- Department of Epidemiology and Cancer Control, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Javier G. Blanco
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St Jude Children’s Research Hospital, Memphis, Tennessee
| | - Paul Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
17
|
Gigliotti JC, Tin A, Pourafshar S, Cechova S, Wang YT, Sung SSJ, Bodonyi-Kovacs G, Cross JV, Yang G, Nguyen N, Chan F, Rebholz C, Yu B, Grove ML, Grams ME, Köttgen A, Scharpf R, Ruiz P, Boerwinkle E, Coresh J, Le TH. GSTM1 Deletion Exaggerates Kidney Injury in Experimental Mouse Models and Confers the Protective Effect of Cruciferous Vegetables in Mice and Humans. J Am Soc Nephrol 2020; 31:102-116. [PMID: 31727850 PMCID: PMC6935006 DOI: 10.1681/asn.2019050449] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 09/07/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND GSTM1 encodes glutathione S-transferase μ-1 (GSTM1), which belongs to a superfamily of phase 2 antioxidant enzymes. The highly prevalent GSTM1 deletion variant is associated with kidney disease progression in human cohorts: the African American Study of Kidney Disease and Hypertension and the Atherosclerosis Risk in Communities (ARIC) Study. METHODS We generated a Gstm1 knockout mouse line to study its role in a CKD model (involving subtotal nephrectomy) and a hypertension model (induced by angiotensin II). We examined the effect of intake of cruciferous vegetables and GSTM1 genotypes on kidney disease in mice as well as in human ARIC study participants. We also examined the importance of superoxide in the mediating pathways and of hematopoietic GSTM1 on renal inflammation. RESULTS Gstm1 knockout mice displayed increased oxidative stress, kidney injury, and inflammation in both models. The central mechanism for kidney injury is likely mediated by oxidative stress, because treatment with Tempol, an superoxide dismutase mimetic, rescued kidney injury in knockout mice without lowering BP. Bone marrow crosstransplantation revealed that Gstm1 deletion in the parenchyma, and not in bone marrow-derived cells, drives renal inflammation. Furthermore, supplementation with cruciferous broccoli powder rich in the precursor to antioxidant-activating sulforaphane significantly ameliorated kidney injury in Gstm1 knockout, but not wild-type mice. Similarly, among humans (ARIC study participants), high consumption of cruciferous vegetables was associated with fewer kidney failure events compared with low consumption, but this association was observed primarily in participants homozygous for the GSTM1 deletion variant. CONCLUSIONS Our data support a role for the GSTM1 enzyme in the modulation of oxidative stress, inflammation, and protective metabolites in CKD.
Collapse
Affiliation(s)
| | - Adrienne Tin
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | | | | | - Yves T Wang
- Division of Nephrology, Department of Medicine, University of Rochester School of Medicine, Rochester, New York
| | | | | | - Janet V Cross
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, Virginia
| | - Guang Yang
- Division of Nephrology, Heinrich-Heine University of Dusseldorf, Dusseldorf, Germany
| | - Nhu Nguyen
- Department of Biomedical Sciences, Grand Valley State University, Allendale, Michigan
| | - Fang Chan
- Division of Nephrology, Department of Medicine and
| | - Casey Rebholz
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health and
| | - Megan L Grove
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Morgan E Grams
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
- Department of Medicine and
| | - Anna Köttgen
- Department of Epidemiology and
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany; and
| | - Robert Scharpf
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland
- Division of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Phillip Ruiz
- Department of Pathology, University of Miami, Miami, Florida
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas
| | - Josef Coresh
- Department of Epidemiology and
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland
| | - Thu H Le
- Division of Nephrology, Department of Medicine and
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Heath, Baltimore, Maryland
| |
Collapse
|
18
|
Copy number variation profiling in pharmacogenes using panel-based exome resequencing and correlation to human liver expression. Hum Genet 2019; 139:137-149. [PMID: 31786673 DOI: 10.1007/s00439-019-02093-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/23/2019] [Indexed: 12/13/2022]
Abstract
Structural variants including copy number variations (CNV) have gained widespread attention, especially in pharmacogenomics but for several genes functional relevance and clinical evidence are still lacking. Detection of CNVs in next-generation sequencing data is challenging but offers widespread applications. We developed a cohort-based CNV detection workflow to extract CNVs from read counts of targeted NGS of 340 genes involved in absorption, distribution, metabolism and excretion (ADME) of drugs. We applied our method to 150 human liver tissue samples and correlated identified CNVs to mRNA expression levels. In total, we identified 445 deletions (73%) and 167 duplications (27%) in 36 pharmacogenes including all well-known CNVs of CYPs, GSTs, SULTs, UGTs, numerous described rare CNVs of CYP2E1, SLC16A3 or UGT2B15 as well as novel observations, e.g., for SLC22A12, SLC22A17 and GPS2 (G Protein Pathway Suppressor 2). We were able to fine-map complex CNVs of CYP2A6 and CYP2D6 with exon resolution. Correlation analysis confirmed known expression patterns for common CNVs and suggested an influence on expression variability for some rare CNVs. Our straightforward CNV detection workflow can be easily applied to any NGS coverage data and helped to analyze CNVs in an ADME-NGS panel of 340 pharmacogenes to improve genotype-phenotype correlations.
Collapse
|
19
|
An Evolutionary Perspective on the Impact of Genomic Copy Number Variation on Human Health. J Mol Evol 2019; 88:104-119. [PMID: 31522275 DOI: 10.1007/s00239-019-09911-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023]
Abstract
Copy number variants (CNVs), deletions and duplications of segments of DNA, account for at least five times more variable base pairs in humans than single-nucleotide variants. Several common CNVs were shown to change coding and regulatory sequences and thus dramatically affect adaptive phenotypes involving immunity, perception, metabolism, skin structure, among others. Some of these CNVs were also associated with susceptibility to cancer, infection, and metabolic disorders. These observations raise the possibility that CNVs are a primary contributor to human phenotypic variation and consequently evolve under selective pressures. Indeed, locus-specific haplotype-level analyses revealed signatures of natural selection on several CNVs. However, more traditional tests of selection which are often applied to single-nucleotide variation often have diminished statistical power when applied to CNVs because they often do not show strong linkage disequilibrium with nearby variants. Recombination-based formation mechanisms of CNVs lead to frequent recurrence and gene conversion events, breaking the linkage disequilibrium involving CNVs. Similar methodological challenges also prevent routine genome-wide association studies to adequately investigate the impact of CNVs on heritable human disease. Thus, we argue that the full relevance of CNVs to human health and evolution is yet to be elucidated. We further argue that a holistic investigation of formation mechanisms within an evolutionary framework would provide a powerful framework to understand the functional and biomedical impact of CNVs. In this paper, we review several cases where studies reveal diverse evolutionary histories and unexpected functional consequences of CNVs. We hope that this review will encourage further work on CNVs by both evolutionary and medical geneticists.
Collapse
|
20
|
Sophonnithiprasert T, Saelee P, Pongtheerat T. GSTM1 and GSTT1 copy number variants and the risk to Thai females of hepatocellular carcinoma. J Gastrointest Oncol 2019; 10:324-329. [PMID: 31032101 DOI: 10.21037/jgo.2018.09.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignancy found throughout the world that most often occurs in males. The cancer is associated with many risk factors such as viral infection, cirrhosis, alcohol, smoking, and fungal toxins. GSTM1 and GSTT1 are detoxification enzymes activated by the cleansing of carcinogenic compounds. Low DNA copy numbers of Glutathione S-transferases M1 and T1 result in a loss of enzyme activity, which causes carcinogenesis factors. DNA copy number variants (CNVs) were determined to compare the differences between the frequencies of GSTM1 and GSTT1 in a control group and patients. Then, the association of these genes with the pathological/survival status of HCC patients was investigated. Methods Forty-nine Thai HCC patients' DNA and the genomic DNA of 66 healthy controls were investigated for GSTM1 and GSTT1 CNVs by real-time polymerase chain reaction (PCR). Then, the correlations between GSTM1 and GSTT1 patients' CNVs, the control group, and clinico-pathological parameters were determined. Results The results show that were no differences between the CNVs of GSTM1 and GSTT1 in the controls and patients (P≥0.05). Only GSTT1 genotypes 0/0 correlated to an increase in the risk of hepatocellular carcinogenesis (OR value was 1.88). GSTM1 CNVs were associated with the gender of patients (P=0.002). However, no correlations were found between GSTT1 CNVs and any of the clinico-pathological parameters. Conclusions The results suggest that only GSTT1 CNVs are associated with increased risk factors of HCC in Thais. GSTM1 copy numbers had a dominant correlation with female HCC patients.
Collapse
Affiliation(s)
- Thanet Sophonnithiprasert
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| | - Pensri Saelee
- Research Division, National Cancer Institute, Bangkok, Thailand
| | - Tanett Pongtheerat
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| |
Collapse
|
21
|
Complex Haplotypes of GSTM1 Gene Deletions Harbor Signatures of a Selective Sweep in East Asian Populations. G3-GENES GENOMES GENETICS 2018; 8:2953-2966. [PMID: 30061374 PMCID: PMC6118300 DOI: 10.1534/g3.118.200462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The deletion of the metabolizing Glutathione S-transferase Mu 1 (GSTM1) gene has been associated with multiple cancers, metabolic and autoimmune disorders, as well as drug response. It is unusually common, with allele frequency reaching up to 75% in some human populations. Such high allele frequency of a derived allele with apparent impact on an otherwise conserved gene is a rare phenomenon. To investigate the evolutionary history of this locus, we analyzed 310 genomes using population genetics tools. Our analysis revealed a surprising lack of linkage disequilibrium between the deletion and the flanking single nucleotide variants in this locus. Tests that measure extended homozygosity and rapid change in allele frequency revealed signatures of an incomplete sweep in the locus. Using empirical approaches, we identified the Tanuki haplogroup, which carries the GSTM1 deletion and is found in approximately 70% of East Asian chromosomes. This haplogroup has rapidly increased in frequency in East Asian populations, contributing to a high population differentiation among continental human groups. We showed that extended homozygosity and population differentiation for this haplogroup is incompatible with simulated neutral expectations in East Asian populations. In parallel, we found that the Tanuki haplogroup is significantly associated with the expression levels of other GSTM genes. Collectively, our results suggest that standing variation in this locus has likely undergone an incomplete sweep in East Asia with regulatory impact on multiple GSTM genes. Our study provides the necessary framework for further studies to elucidate the evolutionary reasons that maintain disease-susceptibility variants in the GSTM1 locus.
Collapse
|
22
|
Oestrogen receptor-regulated glutathione S-transferase mu 3 expression attenuates hydrogen peroxide-induced cytotoxicity, which confers tamoxifen resistance on breast cancer cells. Breast Cancer Res Treat 2018; 172:45-59. [PMID: 30054830 DOI: 10.1007/s10549-018-4897-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 07/17/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Glutathione S-transferase mu 3 (GSTM3) is an enzyme involving in the detoxification of electrophilic compounds by conjugation with glutathione. Higher GSTM3 mRNA levels were reported in patients with ERα-positive breast cancer who received only tamoxifen therapy after surgery. Thus, this study aimed to clarify the oncogenic characteristics of GSTM3 in breast cancer and the mechanism of tamoxifen resistance. METHODS GSTM3 expression in human breast tumour tissues (n = 227) was analysed by RT-PCR and quantitative PCR. Western blot, promoter activity assays, and chromatin immunoprecipitation (ChIP) assays were used to investigate the mechanism of GSTM3 gene regulation. Hydrogen peroxide (H2O2)-induced cytotoxicity in breast cancer cells was detected by MTT assays and flow cytometry. The oncogenic characteristics of GSTM3 in MCF-7 cells were examined by siRNA knockdown in soft agar assays and a xenograft animal model. RESULTS GSTM3 mRNA was highly expressed in ER- and HER2-positive breast cancers. Moreover, patients who received adjuvant Herceptin had increased GSTM3 mRNA levels in tumour tissue. Oestrogen-activated GSTM3 gene expression through ERα-mediated recruitment of SP1, EP300, and AP-1 complexes. GSTM3-silenced MCF-7 cells were more sensitive to H2O2, with significantly inhibited proliferation and colony formation abilities. Tamoxifen-resistant (Tam-R) cells lacking GSTM3 showed enhanced sensitivity to H2O2, but this result was contrary to that obtained after short-term tamoxifen exposure. The animal model suggested that GSTM3 silencing might suppress the tumourigenic ability of MCF-7 cells and increase tumour cell apoptosis. CONCLUSIONS ROS production is one mechanism by which cancer drugs kill tumour cells, and according to our evidence, GSTM3 may play an important role in preventing breast cancer treatment-induced cellular cytotoxicity.
Collapse
|
23
|
Kapahtia S, Hazam RK, Asim M, Karra VK, Chowdhury SJ, Das BC, Kar P. Role of Glutathione S Transferase M1 and T1 Gene Polymorphism in Hepatitis B Related Liver Diseases and Cryptogenic Cirrhosis. J Clin Exp Hepatol 2018; 8:169-172. [PMID: 29892180 PMCID: PMC5992258 DOI: 10.1016/j.jceh.2017.05.208] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Accepted: 05/24/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND AIM Progression of hepatitis B virus infection (HBV) might be affected by host genetic factors. The present study was undertaken to study the role of glutathione S-transferases (GST)-M1 and T1 gene polymorphisms in different stages of HBV infection: HBV inactive carrier, chronic hepatitis B and cirrhosis, and cryptogenic cirrhosis. METHODS The study population comprised of 170 subjects; 120 cases (HBV inactive carrier, n = 30; HBV related chronic hepatitis, n = 30; HBV related cirrhosis, n = 30; cryptogenic cirrhosis, n = 30) and 50 unrelated healthy adults without liver disease as controls. Analysis of GSTM1 and GSTT1 gene polymorphisms was done by multiplex polymerase chain reaction. RESULTS The GSTM1 null genotype was seen more commonly in hepatitis B cirrhosis (n = 21; 70%), chronic hepatitis B (n = 19; 63.33%) and cryptogenic cirrhosis (n = 17; 56.67%) as compared with inactive carrier (n = 9; 30%) and controls (n = 13; 26%). The GSTT1 null genotype was seen less frequently in all the groups, the observed frequencies were controls (n = 7; 14%), inactive carrier (n = 5; 16.67%), chronic hepatitis B (n = 8; 26.67%) and hepatitis B cirrhosis (n = 7; 23.33%). The difference of GSTM1 null genotype frequencies was statistically significant for hepatitis B cirrhosis vs. controls (P = 0.0002), chronic hepatitis B vs. controls (P = 0.002) and cryptogenic cirrhosis vs. controls (P = 0.01). The GSTT1 null genotype was not found to vary significantly between the groups. CONCLUSION The patients with GSTM1 null genotype are at risk of progression of liver disease as the frequency of GSTM1 null genotype was found to be significantly higher in chronic hepatitis B, hepatitis B cirrhosis and cryptogenic cirrhosis as compared with controls.
Collapse
Affiliation(s)
- Siddharth Kapahtia
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India
| | - Rajib K. Hazam
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India
| | - Mohammad Asim
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India
| | - Vijay K. Karra
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India
| | - Soumya J. Chowdhury
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India
| | - Bhudev C. Das
- Dr B.R.Ambedkar Center for Biomedical Research, University of Delhi, India
| | - Premashis Kar
- Department of Medicine, Maulana Azad Medical College, University of Delhi, India,Address for correspondence: Premashis Kar, Department of Medicine, B.L.Taneja Block, R/N-127, New Delhi 110002, India. Tel.: +91 011 23230132; fax: +91 011 23230132.
| |
Collapse
|
24
|
Drug metabolizing enzymes and their inhibitors' role in cancer resistance. Biomed Pharmacother 2018; 105:53-65. [PMID: 29843045 DOI: 10.1016/j.biopha.2018.05.117] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/21/2018] [Accepted: 05/23/2018] [Indexed: 12/15/2022] Open
Abstract
Despite continuous research on chemotherapeutic agents, different mechanisms of resistance have become a major pitfall in cancer chemotherapy. Although, exhaustive efforts are being made by several researchers to target resistance against chemotherapeutic agents, there is another class of resistance mechanism which is almost carrying on unattended. This class of resistance includes pharmacokinetics resistance such as efflux by ABC transporters and drug metabolizing enzymes. ABC transporters are the membrane bound proteins which are responsible for the movement of substrates through the cell membrane. Drug metabolizing enzymes are an integral part of phase-II metabolism that helps in the detoxification of exogenous, endogenous and xenobiotics substrates. These include uridine diphospho-glucuronosyltransferases (UGTs), glutathione-S-transferases (GSTs), dihydropyrimidine dehydrogenases (DPDs) and thiopurine methyltransferases (TPMTs). These enzymes may affect the role of drugs in both positive as well negative manner, depending upon the type of tissue and cells present and when present in tumors, can result in drug resistance. However, the underlying mechanism of resistance by drug metabolizing enzymes is still not clear. Here, we have tried to cover various aspects of these enzymes in relation to anticancer drugs.
Collapse
|
25
|
Saitou M, Satta Y, Gokcumen O, Ishida T. Complex evolution of the GSTM gene family involves sharing of GSTM1 deletion polymorphism in humans and chimpanzees. BMC Genomics 2018; 19:293. [PMID: 29695243 PMCID: PMC5918908 DOI: 10.1186/s12864-018-4676-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 04/15/2018] [Indexed: 02/06/2023] Open
Abstract
Background The common deletion of the glutathione S-transferase Mu 1 (GSTM1) gene in humans has been shown to be involved in xenobiotic metabolism and associated with bladder cancer. However, the evolution of this deletion has not been investigated. Results In this study, we conducted comparative analyses of primate genomes. We demonstrated that the GSTM gene family has evolved through multiple structural variations, involving gene duplications, losses, large inversions and gene conversions. We further showed experimentally that the GSTM1 was polymorphically deleted in both humans and also in chimpanzees, through independent deletion events. To generalize our results, we searched for genic deletions that are polymorphic in both humans and chimpanzees. Consequently, we found only two such deletions among the thousands that we have searched, one of them being the GSTM1 deletion and the other surprisingly being another metabolizing gene, the UGT2B17. Conclusions Overall, our results support the emerging notion that metabolizing gene families, such as the GSTM, NAT, UGT and CYP, have been evolving rapidly through gene duplication and deletion events in primates, leading to complex structural variation within and among species with unknown evolutionary consequences. Electronic supplementary material The online version of this article (10.1186/s12864-018-4676-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Saitou
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, USA
| | - Y Satta
- The Graduate University for Advanced Studies (SOKENDAI), Hayama, Japan
| | - O Gokcumen
- Department of Biological Sciences, State University of New York at Buffalo, Buffalo, USA.
| | - T Ishida
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
26
|
Anderson SM, Naidoo RN, Ramkaran P, Phulukdaree A, Muttoo S, Asharam K, Chuturgoon AA. The Effect of Nitric Oxide Pollution on Oxidative Stress in Pregnant Women Living in Durban, South Africa. ARCHIVES OF ENVIRONMENTAL CONTAMINATION AND TOXICOLOGY 2018; 74:228-239. [PMID: 29063946 DOI: 10.1007/s00244-017-0465-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 09/30/2017] [Indexed: 06/07/2023]
Abstract
The purpose of the study was to evaluate the effect nitric oxide (NO x ) pollution had on maternal serum 8-oxo-7,8-dihydro-2'-deoxyguanosine (8-OHdG) levels and neonatal outcomes in pregnant women living in Durban, South Africa (SA). Women, in their third trimester with singleton pregnancies, were recruited from the heavily industrialised south (n = 225) and less industrialised north (n = 152). Biomarker levels of serum 8-OHdG concentrations were analysed, and the women were genotyped for glutathione-S-transferases pi 1 (GSTP1) and glutathione-S-transferases mu 1 (GSTM1) polymorphisms. The level of NO x pollution in the two regions was determined by using land use regression modelling. The serum 8-OHdG was shown to correlate significantly with NO x levels; this relationship was strengthened in the south (p < 0.05). This relationship was still observed after adjusting for maternal characteristics. GSTP1 was significantly associated with the south region, where the variant (AG+GG) genotype was associated with increased 8-OHdG levels as a result of NO x exposure (p < 0.05). GSTM1 null genotype was associated with a positive correlation between NO x and 8-OHdG levels (p < 0.05). NO x levels were found marginally to reduce gestational age (p < 0.05) with mothers carrying male neonates. Variant GSTP1 and living in the north were factors that contributed to gestational age reduction (p < 0.05). Our study demonstrated that NO x exposure resulted in increased 8-OHdG levels in pregnant women living in Durban, SA, which led to gestational age reduction. The GSTP1 variant increased susceptibility of individuals to harmful effects of NO x .
Collapse
Affiliation(s)
- Samantha M Anderson
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Third Floor, George Campbell Building, Howard Campus, King George V Avenue, Durban, 4041, South Africa
| | - Rajen N Naidoo
- Discipline of Occupational and Environmental Health, School of Nursing and Public Health, College of Health Science, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Prithiksha Ramkaran
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Third Floor, George Campbell Building, Howard Campus, King George V Avenue, Durban, 4041, South Africa
| | - Alisa Phulukdaree
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Third Floor, George Campbell Building, Howard Campus, King George V Avenue, Durban, 4041, South Africa
| | - Sheena Muttoo
- Discipline of Occupational and Environmental Health, School of Nursing and Public Health, College of Health Science, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Kareshma Asharam
- Discipline of Occupational and Environmental Health, School of Nursing and Public Health, College of Health Science, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Third Floor, George Campbell Building, Howard Campus, King George V Avenue, Durban, 4041, South Africa.
| |
Collapse
|
27
|
Girault I, Lidereau R, Bièche I. Trimodal GSTT1 and GSTM1 Genotyping Assay by Real-Time PCR. Int J Biol Markers 2018. [DOI: 10.1177/172460080502000201] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The GSTT1 and GSTM1 genes are characterized by the existence of a GST*0 null allele responsible for a lack of enzyme activity, with the respective null genotypes GSTT1*0/0 and GSTM1*0/0. The three resulting genotypes (GSTs*1/1, *1/0 and *0/0) are associated with a trimodal distribution of glutathione-conjugator activity. Previous epidemiological studies have only evaluated the cancer risk associated with the GST null genotype relative to the two GST carrier geno-® types (GSTs1*1/1 and *1/0). We developed GSTT1 and GSTM1 TaqMan real-time quantitative PCR assays to discriminate each of the three genotypes, with the albumin gene (ALB) as reference. The mean NGSTT1*1/1 value was 1.0 (95% confidence interval 0.80–1.20). The mean NGSTT1*1/0 value was 0.48 (95% CI 0.36–0.60). One (3.4%) of the 29 DNA samples yielded the GSTM1*1/1 genotype (NGSTM1*1/1 = 1), a frequency in keeping with the Hardy-Weinberg distribution. The mean NGSTM1*1/0 value was 0.50 (95% CI 0.42–0.58). All GSTT1*0/0 and GSTM1*0/0 samples yielded NGST values of 0 (Ct = 40); the frequencies of these genotypes (27.6% and 55.2%, respectively) were in keeping with published data. The GSTT1 and GSTM1 real-time PCR assays described here unambiguously discriminate each of the three existing genotypes which should be valuable for assessing the relative risk of cancer associated with each of the three GST genotypes.
Collapse
Affiliation(s)
- I. Girault
- Laboratoire d'Oncogénétique, INSERM
U735, Centre René Huguenin, St-Cloud
| | - R. Lidereau
- Laboratoire d'Oncogénétique, INSERM
U735, Centre René Huguenin, St-Cloud
| | - I. Bièche
- Laboratoire d'Oncogénétique, INSERM
U735, Centre René Huguenin, St-Cloud
- Laboratoire de Génétique Moléculaire,
UPRES EA 3618, Faculté des Sciences Pharmaceutiques et Biologiques, Université René
Descartes, Paris - France
| |
Collapse
|
28
|
Palma-Cano LE, Córdova EJ, Orozco L, Martínez-Hernández A, Cid M, Leal-Berumen I, Licón-Trillo A, Lechuga-Valles R, González-Ponce M, González-Rodríguez E, Moreno-Brito V. GSTT1 and GSTM1 null variants in Mestizo and Amerindian populations from northwestern Mexico and a literature review. Genet Mol Biol 2017; 40:727-735. [PMID: 29111561 PMCID: PMC5738617 DOI: 10.1590/1678-4685-gmb-2016-0142] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 04/05/2017] [Indexed: 12/21/2022] Open
Abstract
The GSTT1 and GSTM1 genes are key molecules in
cellular detoxification. Null variants in these genes are associated with
increase susceptibility to developing different types of cancers. The aim of
this study was to determine the prevalence of GSTT1 and
GSTM1 null genotypes in Mestizo and Amerindian individuals
from the Northwestern region of Mexico, and to compare them with those reported
worldwide. GSTT1 and GSTM1 null variants were
genotyped by multiplex PCR in 211 Mestizos and 211 Amerindian individuals.
Studies reporting on frequency of GSTT1 and
GSTM1 null variants worldwide were identified by a PubMed
search and their geographic distribution were analyzed. We found no significant
differences in the frequency of the null genotype for GSTT1 and
GSM1 genes between Mestizo and Amerindian individuals.
Worldwide frequencies of the GSTT1 and GSTM1
null genotypes ranges from 0.10 to 0.51, and from 0.11 to 0.67, respectively.
Interestingly, in most countries the frequency of the GSTT1
null genotype is common or frequent (76%), whereas the frequency of the
GSMT1 null genotype is very frequent or extremely frequent
(86%). Thus, ethnic-dependent differences in the prevalence of
GSTT1 and GSTM1 null variants may
influence the effect of environmental carcinogens in cancer risk.
Collapse
Affiliation(s)
- Luz Elena Palma-Cano
- Department of Biochemistry, Faculty of Medicine and Biomedical Science, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Emilio J Córdova
- Department of Clinical Research, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Lorena Orozco
- Department of Clinical Research, National Institute of Genomic Medicine, Mexico City, Mexico
| | | | - Miguel Cid
- Department of Clinical Research, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Irene Leal-Berumen
- Department of Biochemistry, Faculty of Medicine and Biomedical Science, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Angel Licón-Trillo
- Department of Biochemistry, Faculty of Medicine and Biomedical Science, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Ruth Lechuga-Valles
- Department of Molecular Biology, Faculty of Zootechnics and Ecology, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Mauricio González-Ponce
- Department of Biochemistry, Faculty of Medicine and Biomedical Science, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Everardo González-Rodríguez
- Department of Molecular Biology, Faculty of Zootechnics and Ecology, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| | - Verónica Moreno-Brito
- Department of Biochemistry, Faculty of Medicine and Biomedical Science, Autonomus University of Chihuahua, Chihuahua, Chihuahua, Mexico
| |
Collapse
|
29
|
Abstract
OBJECTIVE Proteins involving absorption, distribution, metabolism, and excretion (ADME) play a critical role in drug pharmacokinetics. The type and frequency of genetic variation in the ADME genes differ among populations. The aim of this study was to systematically investigate common and rare ADME coding variation in diverse ethnic populations by exome sequencing. MATERIALS AND METHODS Data derived from commercial exome capture arrays and next-generation sequencing were used to characterize coding variation in 298 ADME genes in 251 Northeast Asians and 1181 individuals from the 1000 Genomes Project. RESULTS Approximately 75% of the ADME coding sequence was captured at high quality across the joint samples harboring more than 8000 variants, with 49% of individuals carrying at least one 'knockout' allele. ADME genes carried 50% more nonsynonymous variation than non-ADME genes (P=8.2×10) and showed significantly greater levels of population differentiation (P=7.6×10). Out of the 2135 variants identified that were predicted to be deleterious, 633 were not on commercially available ADME or general-purpose genotyping arrays. Forty deleterious variants within important ADME genes, with frequencies of at least 2% in at least one population, were identified as candidates for future pharmacogenetic studies. CONCLUSION Exome sequencing was effective in accurately genotyping most ADME variants important for pharmacogenetic research, in addition to identifying rare or potentially de novo coding variants that may be clinically meaningful. Furthermore, as a class, ADME genes are more variable and less sensitive to purifying selection than non-ADME genes.
Collapse
|
30
|
Malik MA, Gupta V, Shukla S, Kaur J. Glutathione S-transferase (GSTM1, GSTT1) polymorphisms and JOAG susceptibility: A case control study and meta-analysis in glaucoma. Gene 2017; 628:246-252. [PMID: 28710033 DOI: 10.1016/j.gene.2017.07.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 07/01/2017] [Accepted: 07/10/2017] [Indexed: 10/19/2022]
Abstract
PURPOSE Glutathione S transferase (GST) polymorphisms have been considered risk factors for the development of glaucoma. The aim of the present study was to investigate the association of glutathione S-transferase GSTT1 and GSTM1 genotypes with juvenile open-angle glaucoma (JOAG) in Indian patients. METHODS A case-control study was performed to investigate the associations of GSTM1 and GSTT1 in juvenile open-angle glaucoma. The genotype of GSTM1 and GSTT1 were determined in 73 juvenile open-angle glaucoma patients, and 70 controls matched by age and sex by polymerase chain reaction method. We also performed a meta-analysis of sixteen published studies on GSTM1 and GSTT1 and evaluated the association between the GSTM1 and GSTT1 polymorphisms and glaucoma (JOAG & POAG). Published literature from PubMed and other databases were retrieved. All studies evaluating the association between GSTM1 and GSTT1 polymorphisms and glaucoma (JOAG & POAG) risk were included. Pooled odds ratio (OR) and 95% confidence interval (CI) were calculated using random- or fixed-effects model. RESULTS In the present study, we observed there is no association of GSTM1 (OR=0.680; 95% CI=0.323-1.433; p=0.311) or GSTT1 (OR=0.698; 95% CI=0.307-1.586; p=0.391) with JOAG. In the present meta-analysis, significantly increased glaucoma (JOAG & POAG) risk was found among subjects carrying GSTM1 null genotype (OR=1.177; 95% CI=1.028-1.348; p=0.018) but not among subjects carrying GSTT1 deletion genotype (OR=1.186; 95% CI=0.992-1.417; p=0.061). CONCLUSIONS The present case-control study found that GSTM1 and GSTT1 polymorphism are not associated with JOAG risk in North Indian population. The present meta-analysis suggested that there might be a significant association of GSTM1 null genotype with glaucoma (JOAG & POAG) risk. To the best of our knowledge, this is the first study in the world to investigate role of GSTM1 and GSTT1 polymorphisms with JOAG susceptibility. Given the limited sample size, the associations between GST polymorphism and glaucoma risk needs further investigation.
Collapse
Affiliation(s)
- Manzoor Ahmad Malik
- Department of Ocular Biochemistry, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India; Cancer Diagnostic and Research Centre, Department of Immunology and Molecular Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Soura, Srinagar, J&K 190011, India
| | - Viney Gupta
- Glaucoma Research Facility and Clinical Services, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Shukla
- Department of Ocular Biochemistry, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India
| | - Jasbir Kaur
- Department of Ocular Biochemistry, Dr. Rajendra Prasad Centre for Ophthalmic Sciences, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
31
|
Dastjerdi AH, Behboudi H, Kianmehr Z, Taravati A, Naghizadeh MM, Kaboudanian Ardestani S, Ghazanfari T. Association of glutathione S-transferase polymorphisms with the severity of mustard lung. ACTA ACUST UNITED AC 2017; 7:255-261. [PMID: 29435433 PMCID: PMC5801537 DOI: 10.15171/bi.2017.30] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 08/25/2017] [Accepted: 08/27/2017] [Indexed: 12/25/2022]
Abstract
![]()
Introduction:
Glutathione S-transferase (GST) is one of the major detoxifiers in alveoli. Polymorphism in GST genes can influence the ability of individuals to suppress oxidative stress and inflammation. The present study was aimed to explore the hypothesis that the genetic polymorphisms of GST T1, M1 and P1 are associated with the severity of the mustard lung in the sulfur mustard-exposed individuals.
Methods: Blood samples were taken from 185 sulfur mustard-exposed and 57 unexposed subjects. According to the stage of the mustard lung, sulfur mustard-exposed patients were categorized in the mild/moderate and severe/very severe groups. A multiplex PCR method was conducted to identify GSTM1 and GSTT1 null genotypes. To determine the polymorphisms of GSTP1 in exon 5 (Ile105Val) and exon 6 (Ala114Val), RFLP-PCR method was performed.
Results: The frequency of GSTM1 homozygous deletion was significantly higher in the severe/very severe patients compared with the mild/moderate subjects (66.3% vs. 48%, P = 0.013). The GSTM1 null genotype was associated with the severity of mustard lung (adjusted odds ratio [OR], 2.257; 95% CI, 1.219-4.180). There was no significant association between GSTT1 and GSTP1 polymorphisms with the severity of the mustard lung.
Conclusion: The different distribution of GSTM1 null genotype in severe/very severe and mild/moderate groups indicated that the severity of the mustard lung might be associated with the genetic polymorphism(s).
Collapse
Affiliation(s)
| | - Hossein Behboudi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zahra Kianmehr
- Department of Biology, Faculty of Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ali Taravati
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Mohammad Mehdi Naghizadeh
- Noncommunicable Diseases Research Center, Fasa University of Medical Science, Fasa, Fars Province, Iran
| | | | - Tooba Ghazanfari
- Immunoregulation Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
32
|
Achour Y, Ben Kilani MS, Ben Hamad M, Marzouk S, Mahfoudh N, Bahloul Z, Keskes L, Petit-Teixeira E, Maalej A. Measurement of absolute copy number variation of Glutathione S-Transferase M1 gene by digital droplet PCR and association analysis in Tunisian Rheumatoid Arthritis population. J Clin Lab Anal 2017; 32. [PMID: 28703442 DOI: 10.1002/jcla.22300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 06/15/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The investigation of copy number variations (CNVs) analysis of candidate genes is currently an important research area in modulating human diseases. We aimed to quantify CNVs in glutathione S-transferase M1 (GSTM1) gene and determine its genetic contribution in Tunisian rheumatoid arthritis (RA) and its subsets through an innovative technique for quantification. METHODS A total of 165 RA cases and 102 healthy controls were included in the study. Using a recently powerful approach of digital droplet PCR (ddPCR), we quantified GSTM1 gene to determine the presence of no, one, or multiple copy number (CN) at high levels of sensitivity and specificity. Odds ratio and Fisher exact test were performed to estimate the association risk for GSTM1CNVs in RA. RESULTS Copy number identified by ddPCR was 0, 1, and 2 copies per diploid genome. A high frequency of '0' copy was revealed with 54% in RA patients. The deletion ('0' copy) of GSTM1 was found to be a significant risk factor for anti-cyclic citrullinated peptide (anti-CCP) positive RA (OR=4.16, CI95% =[1.17-14.7]). In addition, a lack of association was found when comparing between the CNVs of RA patients and those of controls. CONCLUSION This study highlights the powerful accuracy of ddPCR for the quantification of CNVs and suggests that the variation in the CN of GSTM1 is associated with anti-CCP positivity in RA. However, it does not indicate a specific role in the susceptibility to the disease in our Tunisian sample.
Collapse
Affiliation(s)
- Yosser Achour
- Laboratory of Human Molecular Genetics, Faculty of Medecine, Sfax, Tunisia
| | | | - Mariem Ben Hamad
- Laboratory of Human Molecular Genetics, Faculty of Medecine, Sfax, Tunisia
| | - Sameh Marzouk
- Department of Internal Medecine, University Hospital Hedi Chaker, Sfax, Tunisia
| | - Nadia Mahfoudh
- Laboratory Services, University Hospital Hedi Chaker, Sfax, Tunisia
| | - Zouheir Bahloul
- Department of Internal Medecine, University Hospital Hedi Chaker, Sfax, Tunisia
| | - Leila Keskes
- Laboratory of Human Molecular Genetics, Faculty of Medecine, Sfax, Tunisia
| | | | - Abdellatif Maalej
- Laboratory of Human Molecular Genetics, Faculty of Medecine, Sfax, Tunisia
| |
Collapse
|
33
|
Wang ZY, Li HY, Jiang Z, Zhou TB, Drummen GPC. GSTM1 Gene Polymorphism is Implicated in Increased Susceptibility to Prostate Cancer in Caucasians and Asians. Technol Cancer Res Treat 2016; 15:NP69-NP78. [PMID: 26614779 DOI: 10.1177/1533034615617650] [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] [Received: 09/23/2015] [Accepted: 10/22/2015] [Indexed: 11/15/2022] Open
Abstract
Published reports on the relationship between GSTM1 gene polymorphisms and prostate cancer risk are heterogeneous in their conclusions, and the significance of these polymorphisms is still debated. This meta-analysis was performed to attempt to combine comparable studies, thereby increasing sample size and statistical significance in order to obtain a better evaluation of the association between GSTM1 polymorphisms and prostate cancer risk. The association investigations were identified from PubMed, Cochrane Library, and China Biological Medicine Database on March 1, 2014. Forty-three reports were recruited into this meta-analysis that contained data from 6741 patients and 9053 controls. There was a marked association between the GSTM1 null genotype and prostate cancer risk in the overall population (odds ratio = 1.39, 95% confidence interval: 1.21-1.60, P <00001), caucasians (odds ratio = 1.48, 95% confidence interval: 1.23-1.79, P <0001) and Asians (odds ratio = 1.62, 95% confidence interval: 1.16-2.27, P = .005). However, the GSTM1 null genotype was not associated with prostate cancer risk in Africans (odds ratio = 0.77, 95% confidence interval: 0.53-1.13, P = 0.19) and African Americans (odds ratio = 1.00, 95% confidence interval: 0.69-1.45, P = 0.99). In conclusion, GSTM1 null genotype was a risk factor to predict the prostate cancer risk in the overall population, Caucasians, and Asians. Although compelling, limitations inherent to meta-analysis, study design of the individual studies, and most importantly, possible gene-gene and gene-environment interactions, as well as the potential involvement of glutathione S-transferases in multiple cellular processes make drawing definite conclusions difficult.
Collapse
Affiliation(s)
- Zhong-Yang Wang
- Department of Urology Surgery, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hong-Yan Li
- Department of Nephrology, Huadu District People's Hospital, Southern Medical University, Guangzhou, China
| | - Zongpei Jiang
- Department of Nephrology, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tian-Biao Zhou
- Department of Nephrology, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Gregor P C Drummen
- Cellular Stress and Ageing Program, Bionanoscience and Bio-Imaging Program, Bio&Nano-Solutions, Bielefeld, Germany
| |
Collapse
|
34
|
Konig-Greger D, Riechelmann H, Wittich U, Gronau S. Genotype and Phenotype of Glutathione-S-Transferase in Patients with Head and Neck Carcinoma. Otolaryngol Head Neck Surg 2016; 130:718-25. [PMID: 15195058 DOI: 10.1016/j.otohns.2003.10.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE: Association of glutathione-S-transferase M1 (GSTM1) polymorphisms and cancer has been demonstrated. Possible underlying mechanisms and genotype-phenotype correlations are not adequately investigated. The aim of this study was to investigate the influence of the GSTM1-null-genotype on the level of GSTM enzyme concentration and on the enzyme activity of GST in patients with head and neck cancer (HNC). METHODS: We investigated in 83 patients and 91 healthy controls the GSTM1 polymorphisms, GSTM1 protein concentration, GSTM1 protein in tumor tissues, and total GST enzyme activity. RESULTS: Total GST enzyme activity was significantly lower in patients with HNC (208 ± 9 μmol/min1) than in controls (264 ± 11 μmol/min1, P < 0.0001) but did not depend on GSTM1-genotype ( P = 0.1). GSTM protein concentration in null-genotype patients (3.6 ± 2.5 μg/mL, mean ± SE) was significantly lower than in GSTM1 allele carriers (26.7 ± 9.6 μg/ml, P < 0.0001); GSTM protein expression did not depend on GSTM1-genotype ( P > 0.5). CONCLUSION: GST enzyme activity in patients with HNC is suppressed, indicating impaired detoxification capacity of tobacco-smoke-related carcinogens. This suppression is not correlated with the GSTM1-genotype. (Otolaryngol Head Neck Surg 2004;130:718-25.)
Collapse
Affiliation(s)
- Diemut Konig-Greger
- Department of Otorhinolaryngology, Institute of Clinical Chemistry, University of Ulm, Germany
| | | | | | | |
Collapse
|
35
|
Kang TY, El-Sohemy A, Comelis MC, Eny KM, Bae SC. Glutathione S-transferase genotype and risk of systemic lupus erythematosus in Koreans. Lupus 2016; 14:381-4. [PMID: 15934438 DOI: 10.1191/0961203305lu2100oa] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Oxidative stress caused by poor detoxification efficiency of reactive oxygen species (ROS) may play a role in the development of systemic lupus erythematosus (SLE). Glutathione S-transferase (GST) is involved in the detoxification of ROS and genetic polymorphisms of GSTM1, GSTT1 and GSTP1 are associated with altered enzyme activity. The aim of this study was to determine whether GSTM1 (deletion), GSTT1 (deletion) and GSTP1 (Ile105! Val105) polymorphisms are associated with susceptibility to SLE or frequency of clinical manifestations according to the ACR diagnostic criteria. DNA was isolated from blood samples collected from 330 patients with SLE and 270 ageand sex-matched controls. GST genotypes were determined by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. No associations were observed between GSTM1, GSTT1, and GSTP1 genotypes and risk of SLE. Among SLE patients, the GSTM1 null genotype was associated with a lower frequency of hematological disorders ( P = 0.012), and a higher SSA(+)/SSB(2) autoantibody profile ( P = 0.042). Compared to SLE patients with the GSTT1 non-null genotype, those with the GSTT1 null genotype had a lower frequency of discoid rash ( P = 0.018), and nephritis ( P = 0.033). Our findings suggest that genetic polymorphisms of GSTM1, GSTT1, and GSTP1 do not influence the risk of SLE, but a deletion of either GSTM1 or GSTT1 may influence certain clinical manifestations of the disease.
Collapse
Affiliation(s)
- T Y Kang
- Department of Rheumatology, Wonju Christian Hospital, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | | | | | | | | |
Collapse
|
36
|
Khrunin AV, Filippova IN, Aliev AM, Tupitsina TV, Slominsky PA, Limborska SA. GSTM1 copy number variation in the context of single nucleotide polymorphisms in the human GSTM cluster. Mol Cytogenet 2016; 9:30. [PMID: 27099630 PMCID: PMC4837583 DOI: 10.1186/s13039-016-0241-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/01/2016] [Indexed: 02/06/2023] Open
Abstract
Background GSTM1 gene deletion is one of the most known copy number polymorphisms in human genome. It is most likely caused by homologous recombination between the repeats flanking the gene. However, taking into account that the deletion has no crucial effects on human well-being, and the ability of other GSTMs to compensate for the lack of GSTM1, a role for additional factors affecting GSTM1 deletion can be proposed. Our goal was to explore the relationships between GSTM1 deletion polymorphism and single nucleotide polymorphisms (SNPs) in the region of the GSTM cluster that includes GSTM2, GSTM3, GSTM4, and GSTM5 in addition to GSTM1. Results Real-time polymerase chain reaction was used to quantify the number of GSTM1 copies. Fourteen SNPs from the region were tested and their allelic patterns were compared in groups of Russian individuals subdivided according to their GSTM1 deletion genotypes. Linkage disequilibrium-based haplotype analysis showed substantial differences of haplotype frequencies between the groups, especially between individuals with homozygous GSTM1 −/− and +/+ genotypes. Exploration of the results of phasing of GSTM1 and SNP genotypes revealed unequal segregation of GSTM1 + and − alleles at different haplotypes. Conclusions The observed differences in haplotype patterns suggest the potential role of genetic context in GSTM1 deletion frequency (appearance) and in the determination of the deletion-related effects.
Collapse
Affiliation(s)
- Andrey V Khrunin
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| | - Irina N Filippova
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| | - Aydar M Aliev
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| | - Tat'yana V Tupitsina
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| | - Petr A Slominsky
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| | - Svetlana A Limborska
- Department of Molecular Bases of Human Genetics, Institute of Molecular Genetics of Russian Academy of Sciences, Kurchatov sq. 2, Moscow, 123182 Russia
| |
Collapse
|
37
|
Sen A, Cingolani P, Senut MC, Land S, Mercado-Garcia A, Tellez-Rojo MM, Baccarelli AA, Wright RO, Ruden DM. Lead exposure induces changes in 5-hydroxymethylcytosine clusters in CpG islands in human embryonic stem cells and umbilical cord blood. Epigenetics 2016; 10:607-21. [PMID: 26046694 DOI: 10.1080/15592294.2015.1050172] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Prenatal exposure to neurotoxicants such as lead (Pb) may cause stable changes in the DNA methylation (5mC) profile of the fetal genome. However, few studies have examined its effect on the DNA de-methylation pathway, specifically the dynamic changes of the 5-hydroxymethylcytosine (5hmC) profile. Therefore, in this study, we investigate the relationship between Pb exposure and 5mC and 5hmC modifications during early development. To study the changes in the 5hmC profile, we use a novel modification of the Infinium™ HumanMethylation450 assay (Illumina, Inc.), which we named HMeDIP-450K assay, in an in vitro human embryonic stem cell model of Pb exposure. We model Pb exposure-associated 5hmC changes as clusters of correlated, adjacent CpG sites, which are co-responding to Pb. We further extend our study to look at Pb-dependent changes in high density 5hmC regions in umbilical cord blood DNA from 48 mother-infant pairs from the Early Life Exposure in Mexico to Environmental Toxicants (ELEMENT) cohort. For our study, we randomly selected umbilical cord blood from 24 male and 24 female children from the 1st and 4th quartiles of Pb levels. Our data show that Pb-associated changes in the 5hmC and 5mC profiles can be divided into sex-dependent and sex-independent categories. Interestingly, differential 5mC sites are better markers of Pb-associated sex-dependent changes compared to differential 5hmC sites. In this study we identified several 5hmC and 5mC genomic loci, which we believe might have some potential as early biomarkers of prenatal Pb exposure.
Collapse
Key Words
- 5-hydroxymethylcytosine
- 5-methylcytosine
- A-clustering approach for grouping HM450K CpG sites into clusters based on correlations
- Aclust
- CpG island, CpG islands are usually in the promoter and 5' untranslated regions of genes
- CpG islands
- CpG, sites, CG dinucleotide methylation site in the genome corresponding with probes on HM450K
- DMRs, Differentially methylated regions (clusters) identified by GEE
- DhMRs, Differentially Hydroxymethylated regions (clusters) identified by GEE
- ELEMENT, Early Life Exposure in Mexico to Environmental Toxicants
- GEE, Generalized Estimating Equations, used to measure the estimated change in DNA methylation in co-regulated regions of the genome
- HM450K
- HM450K, Infinium Human Methylation 450K BeadChip array from Illumina
- HMeDIP-450K
- HMeDIP-450K, Infinium Human Methylation 450K BeadChip array from Illumina coupled with immunoprecipitation with 5hmC antibodies
- LINE-1, Long Interspersed Element 1, a common retrotransposon in humans
- MRs, Methylation regions (clusters) identified by A-clustering
- clusters
- hMRs, Hydroxymethylated regions (clusters) identified by A-clustering
- lead
- umbilical cord blood
Collapse
Affiliation(s)
- Arko Sen
- a Institute of Environmental Health Sciences; Wayne State University ; Detroit , MI , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Pereira D, Assis J, Gomes M, Nogueira A, Medeiros R. Improvement of a predictive model in ovarian cancer patients submitted to platinum-based chemotherapy: implications of a GST activity profile. Eur J Clin Pharmacol 2016; 72:545-53. [PMID: 26803611 DOI: 10.1007/s00228-016-2015-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
PURPOSE The success of chemotherapy in ovarian cancer (OC) is directly associated with the broad variability in platinum response, with implications in patients survival. This heterogeneous response might result from inter-individual variations in the platinum-detoxification pathway due to the expression of glutathione-S-transferase (GST) enzymes. We hypothesized that GSTM1 and GSTT1 polymorphisms might have an impact as prognostic and predictive determinants for OC. METHODS We conducted a hospital-based study in a cohort of OC patients submitted to platinum-based chemotherapy. GSTM1 and GSTT1 genotypes were determined by multiplex PCR. RESULTS GSTM1-null genotype patients presented a significantly longer 5-year survival and an improved time to progression when compared with GSTM1-wt genotype patients (log-rank test, P = 0.001 and P = 0.013, respectively). Multivariate Cox regression analysis indicates that the inclusion of genetic information regarding GSTM1 polymorphism increased the predictive ability of risk of death after OC platinum-based chemotherapy (c-index from 0.712 to 0.833). Namely, residual disease (HR, 4.90; P = 0.016) and GSTM1-wt genotype emerged as more important predictors of risk of death (HR, 2.29; P = 0.039; P = 0.036 after bootstrap). No similar effect on survival was observed regarding GSTT1 polymorphism, and there were no statistically significant differences between GSTM1 and GSTT1 genotypes and the assessed patients' clinical-pathological characteristics. CONCLUSION GSTM1 polymorphism seems to have an impact in OC prognosis as it predicts a better response to platinum-based chemotherapy and hence an improved survival. The characterization of the GSTM1 genetic profile might be a useful molecular tool and a putative genetic marker for OC clinical outcome.
Collapse
Affiliation(s)
- Deolinda Pereira
- Oncology Department, Portuguese Institute of Oncology, Porto, Portugal.,ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal
| | - Joana Assis
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Porto, Portugal
| | - Mónica Gomes
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal.,Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal
| | - Augusto Nogueira
- Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal.,FMUP, Faculty of Medicine, Porto University, Porto, Portugal.,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal
| | - Rui Medeiros
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, Porto, Portugal. .,Molecular Oncology and Viral Pathology Group-Research Center, Portuguese Institute of Oncology, Edifício Laboratórios. 4° piso, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Research Department, Portuguese League Against Cancer (NRNorte), Porto, Portugal. .,CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, Porto, Portugal.
| |
Collapse
|
39
|
Usarek E, Graboń W, Kaźmierczak B, Barańczyk-Kuźma A. Targeting the expression of glutathione- and sulfate-dependent detoxification enzymes in HepG2 cells by oxygen in minimal and amino acid enriched medium. Exp Mol Pathol 2015; 100:82-6. [PMID: 26599691 DOI: 10.1016/j.yexmp.2015.11.020] [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: 11/04/2015] [Accepted: 11/11/2015] [Indexed: 11/18/2022]
Abstract
Cancer cells exhibit specific metabolism allowing them to survive and proliferate in various oxygen conditions and nutrients' availability. Hepatocytes are highly active metabolically and thus very sensitive to hypoxia. The purpose of the study was to investigate the effect of oxygen on the expression of phase II detoxification enzymes in hepatocellular carcinoma cells (HepG2) cultured in minimal and rich media (with nonessential amino acids and GSH). The cells were cultured at 1% hypoxia, 10% tissue normoxia, and 21% atmospheric normoxia. The total cell count was determined by trypan blue exclusion dye and the expression on mRNA level by RT-PCR. The result indicated that the expression of glutathione-dependent enzymes (GSTA, M, P, and GPX2) was sensitive to oxygen and medium type. At 1% hypoxia the enzyme expression (with the exception of GSTA) was higher in minimal compared to rich medium, whereas at 10% normoxia it was higher in the rich medium. The expression was oxygen-dependent in both types of medium. Among phenol sulfotransferase SULT1A1 was not sensitive to studied factors, whereas the expression of SULT1A3 was depended on oxygen only in minimal medium. It can be concluded that in HepG2 cells, the detoxification by conjugation with glutathione and, to a lower extent with sulfate, may be affected by hypoxia and/or limited nutrients' availability. Besides, because the data obtained at 10% oxygen significantly differ from those at 21%, the comparative studies on hypoxia should be performed in relation to 10% but not 21% oxygen.
Collapse
Affiliation(s)
- Ewa Usarek
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Wojciech Graboń
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Beata Kaźmierczak
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Anna Barańczyk-Kuźma
- Chair and Department of Biochemistry, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland.
| |
Collapse
|
40
|
Yoshida N, Inaoka T, Sultana N, Ahmad SA, Mabuchi A, Shimizu H, Watanabe C. Non-monotonic relationships between arsenic and selenium excretion and its implication on arsenic methylation pattern in a Bangladeshi population. ENVIRONMENTAL RESEARCH 2015; 140:300-307. [PMID: 25885118 DOI: 10.1016/j.envres.2015.03.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Revised: 03/16/2015] [Accepted: 03/19/2015] [Indexed: 06/04/2023]
Abstract
The toxicity of arsenic differs markedly between individuals and populations, which might be related to the metabolism (methylation) of inorganic arsenic (As), as well as the selenium (Se) nutritional status. Urinary excretion of As (u-As) and Se (u-Se) was examined in an adult population (n=128) living in an As-contaminated area in Bangladesh. Although there was a significant negative correlation between u-Se and u-As (median 137; range 49-927 μg/g creatinine), closer examination revealed a non-monotonous relationship between them. A quadratic curve with an axis of As at 155 μg/g Cre gave a better fit, and u-As and u-Se were positively or negatively correlated depending on whether the As concentration was lower or higher than 155 μg As/g Cre, respectively. Likewise, the relationships between the As methylation pattern and glutathione-S-transferase (GST) polymorphism, body mass index (BMI), and u-Se differed depending on the u-As range; i.e., higher or lower than 155 μg/g Cre. Although we did not determine the causal mechanism for these observations, the non-monotonic relationship between As exposure and the variables examined suggested the existence of a threshold at which the handling of As by human body is qualitatively changed. The possible importance of Se nutrition for As toxicity is also discussed.
Collapse
Affiliation(s)
- Nao Yoshida
- Department of Human Ecology, School of International Health, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Tsukasa Inaoka
- Department of Human Ecology, Faculty of Agriculture, Saga University, 1 Honjo-machi, Saga 840-8502, Japan
| | - Nayar Sultana
- Department of Human Ecology, School of International Health, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Sk Akhtar Ahmad
- Department of Occupational and Environmental Medicine, Bangladesh University of Health Sciences, Mirpur, Dhaka 1216, Bangladesh
| | - Akihiko Mabuchi
- Department of Human Genetics, School of International Health, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hana Shimizu
- Department of Human Ecology, School of International Health, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Chiho Watanabe
- Department of Human Ecology, School of International Health, Graduate School of Medicine, The University of Tokyo, Hongo 7-3-1, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
41
|
Roberts RL, Barclay ML. Update on thiopurine pharmacogenetics in inflammatory bowel disease. Pharmacogenomics 2015; 16:891-903. [PMID: 26067482 DOI: 10.2217/pgs.15.29] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Azathioprine and 6-mercaptopurine remain pivotal therapies for the maintenance of disease remission in patients with Crohn's disease and ulcerative colitis. While thiopurine S-methyltransferase deficiency was the first pharmacogenetic phenomenon to be recognized to influence thiopurine toxicity and reliably predict leukopenia, it does not predict other adverse effects, nor does it explain most cases of thiopurine resistance. In recent years, a number of other genetic polymorphisms have received increasing attention in the literature. In particular, SNPs in NUDT15 and in the class II HLA locus have been shown to predict thiopurine-related leukopenia and pancreatitis. The aim of this review is to provide a concise update of genetic variability which may influence patient response to azathioprine and 6-mercaptopurine.
Collapse
Affiliation(s)
- Rebecca L Roberts
- Department of Surgical Sciences, Dunedin School of Medicine, PO Box 56, Dunedin, New Zealand
| | - Murray L Barclay
- Department of Medicine, University of Otago Christchurch, PO Box 4345, Christchurch, New Zealand.,Department of Gastroenterology, Christchurch Hospital, Private Bag 4710, Christchurch, New Zealand
| |
Collapse
|
42
|
Conway K, Edmiston SN, Tse CK, Bryant C, Kuan PF, Hair BY, Parrish EA, May R, Swift-Scanlan T. Racial variation in breast tumor promoter methylation in the Carolina Breast Cancer Study. Cancer Epidemiol Biomarkers Prev 2015; 24:921-30. [PMID: 25809865 PMCID: PMC4452445 DOI: 10.1158/1055-9965.epi-14-1228] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/03/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND African American (AA) women are diagnosed with more advanced breast cancers and have worse survival than white women, but a comprehensive understanding of the basis for this disparity remains unclear. Analysis of DNA methylation, an epigenetic mechanism that can regulate gene expression, could help to explain racial differences in breast tumor clinical biology and outcomes. METHODS DNA methylation was evaluated at 1,287 CpGs in the promoters of cancer-related genes in 517 breast tumors of AA (n = 216) or non-AA (n = 301) cases in the Carolina Breast Cancer Study (CBCS). RESULTS Multivariable linear regression analysis of all tumors, controlling for age, menopausal status, stage, intrinsic subtype, and multiple comparisons [false discovery rate (FDR)], identified seven CpG probes that showed significant (adjusted P < 0.05) differential methylation between AAs and non-AAs. Stratified analyses detected an additional four CpG probes differing by race within hormone receptor-negative (HR(-)) tumors. Genes differentially methylated by race included DSC2, KCNK4, GSTM1, AXL, DNAJC15, HBII-52, TUSC3, and TES; the methylation state of several of these genes may be associated with worse survival in AAs. TCGA breast tumor data confirmed the differential methylation by race and negative correlations with expression for most of these genes. Several loci also showed racial differences in methylation in peripheral blood leukocytes (PBL) from CBCS cases, indicating that these variations were not necessarily tumor-specific. CONCLUSIONS Racial differences in the methylation of cancer-related genes are detectable in both tumors and PBLs from breast cancer cases. IMPACT Epigenetic variation could contribute to differences in breast tumor development and outcomes between AAs and non-AAs.
Collapse
Affiliation(s)
- Kathleen Conway
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| | - Sharon N Edmiston
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Chiu-Kit Tse
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christopher Bryant
- Department of Biostatistics, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Pei Fen Kuan
- Department of Applied Mathematics and Statistics, State University of New York, Stony Brook, New York
| | - Brionna Y Hair
- Department of Epidemiology, School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eloise A Parrish
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Ryan May
- The EMMES Corporation, Rockville, Maryland
| | - Theresa Swift-Scanlan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina. School of Nursing, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
43
|
Beyerle J, Frei E, Stiborova M, Habermann N, Ulrich CM. Biotransformation of xenobiotics in the human colon and rectum and its association with colorectal cancer. Drug Metab Rev 2015; 47:199-221. [PMID: 25686853 DOI: 10.3109/03602532.2014.996649] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In humans, the liver is generally considered to be the major organ contributing to drug metabolism, but studies during the last years have suggested an important role of the extra-hepatic drug metabolism. The gastrointestinal tract (GI-tract) is the major path of entry for a wide variety of compounds including food, and orally administered drugs, but also compounds - with neither nutrient nor other functional value - such as carcinogens. These compounds are metabolized by a large number of enzymes, including the cytochrome P450 (CYP), the glutathione S-transferase (GST) family, the uridine 5'-diphospho- glucuronosyltransferase (UDP-glucuronosyltransferase - UGT) superfamily, alcohol-metabolizing enzymes, sulfotransferases, etc. These enzymes can either inactivate carcinogens or, in some cases, generate reactive species with higher reactivity compared to the original compound. Most data in this field of research originate from animal or in vitro studies, wherein human studies are limited. Here, we review the human studies, in particular the studies on the phenotypic expression of these enzymes in the colon and rectum to get an impression of the actual enzyme levels in this primary organ of exposure. The aim of this review is to give a summary of currently available data on the relation between the CYP, the GST and the UGT biotransformation system and colorectal cancer obtained from clinical and epidemiological studies in humans.
Collapse
Affiliation(s)
- Jolantha Beyerle
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) , Heidelberg , Germany
| | | | | | | | | |
Collapse
|
44
|
Saitou M, Ishida T. Distributions of the GSTM1 and GSTT1 null genotypes worldwide are characterized by latitudinal clines. Asian Pac J Cancer Prev 2015; 16:355-61. [PMID: 25640380 DOI: 10.7314/apjcp.2015.16.1.355] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deletion types of genetic variants of glutathione S-transferase (GST) M1 and T1, the GSTM1 null and GSTT1 null which are risk factors for certain cancers, have been ubiquitously found in human populations but their worldwide distribution pattern is unclear. MATERIALS AND METHODS To perform a meta-analysis, a systematic search for the literature on GSTM1 and GSTT1 null genotypes was done to identify 63 reports for 81 human populations. Relationships between the GSTM1 and GSTT1 null genotype frequencies and the absolute latitude of 81 populations were tested by Spearman's rank correlation coefficient. RESULTS A significant positive correlation was detected between the GSTM1 null genotype frequency and the absolute latitude (r=0.28, p-value <0.05), whereas the GSTT1 null genotype frequency and absolute latitude showed a significant negative correlation (r= -0.41 p-value <0.01). There was no correlation between the frequencies of GSTM1 and GSTT1 null genotype in each population (r= -0.029, p-value=0.80). CONCLUSIONS Latitudinal clines of the distribution of the GSTM1 and GSTT1 null genotypes may be attributed to the result of gene-environmental adaptation. No functional compensation between GSTM1 and GSTT1 was suggested by the lack of correlation between the null frequencies for GSTM1 and GSTT1.
Collapse
Affiliation(s)
- Marie Saitou
- Unit of Human Biology and Genetics, Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan E-mail :
| | | |
Collapse
|
45
|
Emeville E, Broquère C, Brureau L, Ferdinand S, Blanchet P, Multigner L, Romana M. Copy number variation of GSTT1 and GSTM1 and the risk of prostate cancer in a Caribbean population of African descent. PLoS One 2014; 9:e107275. [PMID: 25198353 PMCID: PMC4157893 DOI: 10.1371/journal.pone.0107275] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 08/09/2014] [Indexed: 11/24/2022] Open
Abstract
Background Deletions of the glutathione S-transferase genes M1 and T1 (GSTM1 and GSTT1) have been studied as potential risk factors for prostate cancer. Conflicting results have been obtained. Moreover, most such studies could not discriminate heterozygous from homozygous carriers of the non-deleted alleles. Objective We investigated whether copy number variation (CNV) of the GSTM1 and/or GSTT1 genes contribute to the risk of prostate cancer in the Caribbean population of African descent of Guadeloupe. Methods In a population-based case-control study, we compared 629 prostate cancer patients and 622 control subjects. Logistic regression was used to estimate adjusted odds ratios (OR) and 95% confidence intervals (CI). Exact copy numbers of GSTM1 and GSTT1 were determined by real-time PCR. Results A higher copy number of GSTM1 was marginally associated with prostate cancer risk. Men with 2 and 3 or more GSTT1 genes were at higher risk of prostate cancer (OR: 1.55, 95% CI: 1.11–2.16 and OR: 4.89, 95% CI: 1.71–13.99, respectively; Ptrend<0.001). Men with 3, 4 and 5 or more copies of both GSTM1 and GSTT1 genes were at higher risk of prostate cancer (OR: 2.18, 95% CI: 1.21–3.91, OR: 3.24, 95% CI: 1.63–6.46, and OR: 5.77, 95% CI: 1.40–23.84, respectively; Ptrend<0.001). Conclusions Copy number of GSTT1 and combined GSTM1/GSTT1 appear to be associated with prostate cancer risk in our population study with gene dose relationship. Our results support the hypothesis that variations in copy number of GSTT1 modulate the risk of prostate cancer.
Collapse
Affiliation(s)
- Elise Emeville
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1085, Institut de Recherche sur la Santé, l’Environnement et le Travail (IRSET), Pointe-à-Pitre, Guadeloupe, France
- Université de Rennes 1, Rennes, France
| | - Cédric Broquère
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1134, Pointe-à-Pitre, Guadeloupe, France
- Université des Antilles et de la Guyane, Pointe-à-Pitre, Guadeloupe, France
| | - Laurent Brureau
- Service d’Urologie, Centre Hospitalier Universitaire de Pointe à Pitre, Pointe à Pitre, Guadeloupe, France
| | - Séverine Ferdinand
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1134, Pointe-à-Pitre, Guadeloupe, France
- Université des Antilles et de la Guyane, Pointe-à-Pitre, Guadeloupe, France
| | - Pascal Blanchet
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1085, Institut de Recherche sur la Santé, l’Environnement et le Travail (IRSET), Pointe-à-Pitre, Guadeloupe, France
- Université des Antilles et de la Guyane, Pointe-à-Pitre, Guadeloupe, France
- Service d’Urologie, Centre Hospitalier Universitaire de Pointe à Pitre, Pointe à Pitre, Guadeloupe, France
| | - Luc Multigner
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1085, Institut de Recherche sur la Santé, l’Environnement et le Travail (IRSET), Pointe-à-Pitre, Guadeloupe, France
- Université de Rennes 1, Rennes, France
- * E-mail:
| | - Marc Romana
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1134, Pointe-à-Pitre, Guadeloupe, France
- Université des Antilles et de la Guyane, Pointe-à-Pitre, Guadeloupe, France
| |
Collapse
|
46
|
Bansal VK, Rajan K, Sharma A, Paliwal P, Chaubal G, Jindal V, Misra MC, Kucheria K. Prospective Case-Control Study to Evaluate the Role of Glutathione S Transferases (GSTT1 and GSTM1) Gene Deletion in Breast Carcinoma and Its Prognostic Significance. Indian J Surg 2014; 77:1067-72. [PMID: 27011512 DOI: 10.1007/s12262-014-1152-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 07/22/2014] [Indexed: 11/24/2022] Open
Abstract
Breast cancer is the most common cause of cancer death in women with the incidence rising in young women. GST gene polymorphisms are significant because of their role in the detoxification of both environmental carcinogens and also cytotoxic drugs used in therapy for breast cancer. The present study has been designed to identify the role of polymorphisms in GSTT1 and GSTM1 genes in the risk of development of breast cancer, in the prognostication of breast cancer, and in the prediction of response towards chemotherapy. Ninety-nine patients with breast cancer and 100 healthy controls with no history of cancer were taken from blood donors after informed consent. Epidemiological and clinical data was collected from participants and 5 ml of peripheral venous blood was collected for genotype analysis. Null genotype of GSTT1 was detected in 51.04 % of the controls in comparison to 20.2 % of patients with carcinoma breast, which was found to be statistically significant (OR 4.18; 95 % CI 2.01-8.75; P = 0.0001). GSTM1 gene deletion was also significantly more common among controls (60 %) than in patients with breast cancer (33 %) (OR 4.57; 95 % CI 2.20-9.51; P = 0.0001). Tumors more than 5 cm in size had greater tendency for GSTM1 gene expression (P value = 0.019), but other clinicopathological parameters did not show any correlation. GSTT1 and GSTM1 genes status did not show any association with response to chemotherapy. The results indicated the null genotype of both GSTT1 and GSTM1 to be protective for the development of carcinoma breast. None of the known etiological factors have any correlation with GSTT1 and GSTM1 gene deletion. Patients with small tumor size expressed GSTM1 gene deletion. Other tumor characteristics and clinicopathological parameters did not have any correlation with gene deletion.
Collapse
Affiliation(s)
- Virinder Kumar Bansal
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Room No. 5021, 5th Floor Teaching Block, New Delhi, India
| | - Karthik Rajan
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Room No. 5021, 5th Floor Teaching Block, New Delhi, India
| | - Arundhati Sharma
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Preeti Paliwal
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| | - Gaurav Chaubal
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Room No. 5021, 5th Floor Teaching Block, New Delhi, India
| | - Vikas Jindal
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Room No. 5021, 5th Floor Teaching Block, New Delhi, India
| | - Mahesh C Misra
- Department of Surgical Disciplines, All India Institute of Medical Sciences, Room No. 5021, 5th Floor Teaching Block, New Delhi, India
| | - Kiran Kucheria
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
47
|
Fundia AF, Weich N, Crivelli A, La Motta G, Larripa IB, Slavutsky I. Glutathione S-transferase gene polymorphisms in celiac disease and their correlation with genomic instability phenotype. Clin Res Hepatol Gastroenterol 2014; 38:379-84. [PMID: 24565472 DOI: 10.1016/j.clinre.2014.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/27/2013] [Accepted: 01/14/2014] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND OBJECTIVE Genomic instability and reduced glutathione S-transferase (GST) activity have been identified as potential risk factors for malignant complications in celiac disease (CD). In this study, we assessed the possible influence of GST polymorphisms on genome instability phenotypes in a genetically characterised group of celiac patients from previous studies. METHODS The deletion polymorphisms in GSTM1 and GSTT1 genes and the single-nucleotide polymorphism GSTP1 c.313A>G were genotyped using PCR in a set of 20 untreated adult patients with a known genomic instability phenotype and 69 age- and sex-matched healthy individuals. RESULTS The frequencies of variant genotypes in patients were GSTM1-null (30%), GSTT1-null (5%), GSTP1-AG (60%) and GSTP1-GG (15%), and they showed no differences from controls. No significant differences were found in the genotype distribution based on telomere length. Cases with GSTM1-null genotype (83%) and microsatellite stability were more frequent than those with genomic instability. Moreover, carriers of GSTP1-variant genotype (73%) and stable phenotype were significantly increased compared to unstable patients (27%) (P=0.031). No differences were found according to the clinical-pathological characteristics of celiac cases. CONCLUSIONS No association between GST polymorphic variants and celiac-associated genomic instability was proven in our cohort. Future studies should explore the usefulness of other biomarkers to distinguish celiac patients who are susceptible to cancer development.
Collapse
Affiliation(s)
- Ariela F Fundia
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental (IMEX), CONICET/ANM, Academia Nacional de Medicina, Pacheco de Melo 3081, Buenos Aires C1425AUM, Capital Federal, Argentina.
| | - Natalia Weich
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental (IMEX), CONICET/ANM, Academia Nacional de Medicina, Pacheco de Melo 3081, Buenos Aires C1425AUM, Capital Federal, Argentina
| | - Adriana Crivelli
- Servicio de Soporte Nutricional y Malabsorción, Hospital San Martín, 1900 La Plata, Argentina
| | - Graciela La Motta
- Servicio de Soporte Nutricional y Malabsorción, Hospital San Martín, 1900 La Plata, Argentina
| | - Irene B Larripa
- Laboratorio de Genética Hematológica, Instituto de Medicina Experimental (IMEX), CONICET/ANM, Academia Nacional de Medicina, Pacheco de Melo 3081, Buenos Aires C1425AUM, Capital Federal, Argentina
| | - Irma Slavutsky
- Laboratorio de Genética de Neoplasias Linfoides, Instituto de Medicina Experimental (IMEX), CONICET/ANM, Buenos Aires C1425AUM, Argentina
| |
Collapse
|
48
|
Maniglia MP, Ribeiro MEB, Costa NMD, Jacomini MLG, Carvalho TBOD, Molina FD, Piatto VB, Maniglia JV. Molecular pathogenesis of juvenile nasopharyngeal angiofibroma in brazilian patients. Pediatr Hematol Oncol 2013; 30:616-22. [PMID: 23802802 DOI: 10.3109/08880018.2013.806620] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Juvenile nasopharyngeal angiofibroma (JNA) is a vascular tumor of the nasopharynx that accounts for 0.5% of all cancers of the head and neck. It primarily affects males aged 14-25 years. Of the many genes that mediate the development of JNA, GSTM1 has been most frequently associated with this vascular tumor. The loss of expression of GSTM1 (null genotype) is linked to the development of these tumors. The aim of this cross-sectional case study was to examine the prevalence of the GSTM1-null genotype in Brazilian patients with JNA. DNA was extracted from the leukocytes of blood samples from 10 patients. GSTM1 genotypes were analyzed using a PCR-based assay that was designed to identify the wild-type allele of GSTM1. All 10 patients (100%) were males, with a mean age of 17.8 years. The null genotype for GSTM1 was noted in 4 patients (40%)-1 (10%) at Fisch stage I, 1 (10%) at stage III, and 2 (20%) at stage II. No patient with this genotype had stage IV disease. There was no correlation between Fisch classification and GSTM1 genotype (P = .5695). The correlation between age at diagnosis and GSTM1 genotype was not significant (P = .728). The present findings indicate that there is evidence of an association between the GSTM1-null genotype and JNA in this studied Brazilian population.
Collapse
Affiliation(s)
- Maurício Pereira Maniglia
- 1Department of Otorhinolaryngology and Head-Neck Surgery of the Faculty of Medicine of São José do Rio Preto Medical School (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhang X, Huang M, Wu X, Kadlubar S, Lin J, Yu X, Fan C, Ning B, Kadlubar FF. GSTM1 copy number and promoter haplotype as predictors for risk of recurrence and/or second primary tumor in patients with head and neck cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2013; 6:9-17. [PMID: 23526580 PMCID: PMC3596140 DOI: 10.2147/pgpm.s35949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Indexed: 11/23/2022]
Abstract
The objective of this study was to determine copy number variant (CNV) and promoter genetic variants in glutathione S-transferase Mu class 1 (GSTM1) and the risk of recurrence (REC)/second primary tumor (SPT) in patients with previously diagnosed early stage head and neck cancer. Among 441 subjects, 133 experienced REC and/or an SPT, while 308 had single primary disease. TaqMan real-time polymerase chain reaction was used to measure the exact copy number of GSTM1 and direct sequencing was used to determine genetic variants in the GSTM1 promoter region. Multivariate Cox regression analysis was performed to estimate hazard ratios (HRs) and 95% confidence intervals (95% CIs) associated with copy number and genetic variants. REC/SPT-free survival times were compared by constructing Kaplan–Meier curves and differences between curves were tested by logrank test. Results showed a significantly decreased REC/SPT (HR = 0.57; 95% CI = 0.35–0.95) and longer REC/SPT-free survival in subjects with at least two copies of GSTM1 compared with the GSTM1 homozygous deletion, but not in those with one copy of GSTM1. The −498G, −426G, and −339T alleles were significantly associated with REC/SPT, with HRs of 0.11 (0.02–0.85), 0.28 (0.11–0.74) and 2.02 (1.07–3.82), respectively. Kaplan–Meier survival analysis showed that the −498G, −426G, and −339C alleles were also significantly associated with increased REC/SPT-free survival. Further haplotype analysis showed the haplotype P−498G-−426G-−339C carriers had decreased REC/SPT with a HR of 0.09 (95% CI 0.01–0.71) and increased REC/SPT-free survival compared with those with haplotype P−498C-−426A-−339T. The P−498C-−426A-−339T-containing reporter construct had significantly increased luciferase expression. These results suggest that the GSTM1 CNV and promoter haplotype are better predictors of REC/SPTs of head and neck cancer than just measuring the presence/absence of GSTM1.
Collapse
Affiliation(s)
- Xuemei Zhang
- University of Arkansas for Medical Sciences, Little Rock, Arkansas, US Food and Drug Administration, Jefferson, Arkansas, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hur J, Kim H, Ha EH, Park H, Ha M, Kim Y, Hong YC, Chang N. Birth weight of Korean infants is affected by the interaction of maternal iron intake and GSTM1 polymorphism. J Nutr 2013; 143:67-73. [PMID: 23173169 DOI: 10.3945/jn.112.161638] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Excessive iron consumption during pregnancy can lead to increased oxidative stress in the maternal body, which may result in adverse pregnancy outcomes. Glutathione S-transferases (GSTs) originate from a superfamily of detoxifying enzymes that play a role in reducing xenobiotic compounds and oxidative stress. The aim of this study was to determine the relationship among GST gene expression, maternal iron intake during pregnancy, and neonatal birth weight. The study participants were 1087 Korean gravidas and their newborns recruited for the Mothers and Children's Environmental Health study between 2006 and 2010. A 24-h dietary recall interview was conducted to estimate iron intake; additional intake through nutritional supplements was thoroughly investigated. Deletion polymorphisms of GSTM1 and GSTT1 were genotyped using PCR. Dietary iron consumption during pregnancy was positively associated with birth weight in pregnant women who were GSTM1-present after adjustment for the following covariates: maternal age, prepregnancy BMI, mother's education level, log-transformed urinary cotinine level, infant gender, gestational age at term, log-transformed energy intake, parity, and the use of folic acid supplements (P < 0.05). There were interactions between the GSTM1 genotype and iron intakes from animal foods (P < 0.05), diet (P < 0.05), and diet with supplements (P < 0.05). No relationship was found between maternal iron intake and birth weight for the GSTT1 polymorphism. This study demonstrates that increased iron consumption during pregnancy may improve infant birth weight for mothers who are GSTM1-present, but it might not be beneficial for mothers with the GSTM1-null genotype.
Collapse
Affiliation(s)
- Jinhee Hur
- Department of Nutritional Science and Food Management, College of Medicine, Ewha Womans University, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|