Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732205

ABSTRACT

The tumor microenvironment is affected by reactive oxygen species and has been suggested to have an important role in ovarian cancer (OC) tumorigenesis. The role of glutathione transferases (GSTs) in the maintenance of redox balance is considered as an important contributing factor in cancer, including OC. Furthermore, GSTs are mostly encoded by highly polymorphic genes, which further highlights their potential role in OC, known to originate from accumulated genetic changes. Since the potential relevance of genetic variations in omega-class GSTs (GSTO1 and GSTO2), with somewhat different activities such as thioltransferase and dehydroascorbate reductase activity, has not been clarified as yet in terms of susceptibility to OC, we aimed to investigate whether the presence of different GSTO1 and GSTO2 genetic variants, individually or combined, might represent determinants of risk for OC development. Genotyping was performed in 110 OC patients and 129 matched controls using a PCR-based assay for genotyping single nucleotide polymorphisms. The results of our study show that homozygous carriers of the GSTO2 variant G allele are at an increased risk of OC development in comparison to the carriers of the referent genotype (OR1 = 2.16, 95% CI: 0.88-5.26, p = 0.08; OR2 = 2.49, 95% CI: 0.93-6.61, p = 0.06). Furthermore, individuals with GST omega haplotype H2, meaning the concomitant presence of the GSTO1*A and GSTO2*G alleles, are more susceptible to OC development, while carriers of the H4 (*A*A) haplotype exhibited lower risk of OC when crude and adjusted haplotype analysis was performed (OR1 = 0.29; 95% CI: 0.12-0.70; p = 0.007 and OR2 = 0.27; 95% CI: 0.11-0.67; p = 0.0054). Overall, our results suggest that GSTO locus variants may confer OC risk.


Subject(s)
Alleles , Genetic Predisposition to Disease , Glutathione Transferase , Ovarian Neoplasms , Polymorphism, Single Nucleotide , Humans , Female , Ovarian Neoplasms/genetics , Glutathione Transferase/genetics , Middle Aged , Genotype , Adult , Aged , Case-Control Studies , Gene Frequency
2.
Medicina (Kaunas) ; 60(4)2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38674199

ABSTRACT

Background and Objectives: Despite improvements in screening programs, a large number of patients with colorectal cancer (CRC) are diagnosed in an advanced disease stage. Previous investigations imply that glutathione transferases (GSTs) might be associated with the development and progression of CRC. Moreover, the detoxification mechanism of oxaliplatin, which represents the first line of treatment for advanced CRC, is mediated via certain GSTs. The aim of this study was to evaluate the significance of certain GST genetic variants on CRC prognosis and the efficacy of oxaliplatin-based treatment. Materials and Methods: This prospective study included 523 patients diagnosed with CRC in the period between 2014 and 2016, at the Digestive Surgery Clinic, University Clinical Center of Serbia, Belgrade. Patients were followed for a median of 43.47 ± 17.01 months (minimum 1-63 months). Additionally, 109 patients with advanced disease, after surgical treatment, received FOLFOX6 treatment as a first-line therapy between 2014 and 2020. The Kaplan-Meier method was used to analyze cumulative survival, and the Cox proportional hazard regression model was used to study the effects of different GST genotypes on overall survival. Results: Individuals with the GSTM1-null genotype and the GSTP1 IleVal+ValVal (variant) genotype had significantly shorter survival when compared to referent genotypes (GSTM1-active and GSTP1 IleIle) (log-rank: p = 0.001). Moreover, individuals with the GSTM1-null genotype who received 5-FU-based treatment had statistically significantly shorter survival when compared to individuals with the GSTM1-active genotype (log-rank: p = 0.05). Conclusions: Both GSTM1-null and GSTP1 IleVal+ValVal (variant) genotypes are associated with significantly shorter survival in CRC patients. What is more, the GSTM1-null genotype is associated with shorter survival in patients receiving FOLOFOX6 treatment.


Subject(s)
Adenocarcinoma , Colorectal Neoplasms , Glutathione S-Transferase pi , Glutathione Transferase , Polymorphism, Genetic , Humans , Glutathione S-Transferase pi/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/drug therapy , Glutathione Transferase/genetics , Female , Male , Middle Aged , Aged , Prospective Studies , Serbia , Adenocarcinoma/genetics , Adenocarcinoma/mortality , Adenocarcinoma/drug therapy , Fluorouracil/therapeutic use , Genotype , Prognosis , Kaplan-Meier Estimate , Organoplatinum Compounds/therapeutic use , Leucovorin/therapeutic use , Oxaliplatin/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Proportional Hazards Models , Adult
3.
Int J Mol Sci ; 24(12)2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37373377

ABSTRACT

Although disturbance of redox homeostasis might be responsible for COVID-19 cardiac complications, this molecular mechanism has not been addressed yet. We have proposed modifying the effects of antioxidant proteins polymorphisms (superoxide dismutase 2 (SOD2), glutathione peroxidase 1 (GPX1), glutathione peroxidase 3 (GPX3) and nuclear factor erythroid 2-related factor 2, (Nrf2)) in individual susceptibility towards the development of cardiac manifestations of long COVID-19. The presence of subclinical cardiac dysfunction was assessed via echocardiography and cardiac magnetic resonance imaging in 174 convalescent COVID-19 patients. SOD2, GPX1, GPX3 and Nrf2 polymorphisms were determined via the appropriate PCR methods. No significant association of the investigated polymorphisms with the risk of arrhythmia development was found. However, the carriers of variant GPX1*T, GPX3*C or Nrf2*A alleles were more than twice less prone for dyspnea development in comparison with the carriers of the referent ones. These findings were even more potentiated in the carriers of any two variant alleles of these genes (OR = 0.273, and p = 0.016). The variant GPX alleles were significantly associated with left atrial and right ventricular echocardiographic parameters, specifically LAVI, RFAC and RV-EF (p = 0.025, p = 0.009, and p = 0.007, respectively). Based on the relation between the variant SOD2*T allele and higher levels of LV echocardiographic parameters, EDD, LVMI and GLS, as well as troponin T (p = 0.038), it can be proposed that recovered COVID-19 patients, who are the carriers of this genetic variant, might have subtle left ventricular systolic dysfunction. No significant association between the investigated polymorphisms and cardiac disfunction was observed when cardiac magnetic resonance imaging was performed. Our results on the association between antioxidant genetic variants and long COVID cardiological manifestations highlight the involvement of genetic propensity in both acute and long COVID clinical manifestations.


Subject(s)
Antioxidants , COVID-19 , Humans , Post-Acute COVID-19 Syndrome , NF-E2-Related Factor 2 , COVID-19/diagnostic imaging , COVID-19/genetics , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Glutathione Peroxidase GPX1 , Superoxide Dismutase/metabolism , Echocardiography
4.
Int J Mol Sci ; 24(22)2023 Nov 10.
Article in English | MEDLINE | ID: mdl-38003341

ABSTRACT

In SARS-CoV-2 infection, excessive activation of the immune system intensively increases reactive oxygen species levels, causing harmful hyperinflammatory and oxidative state cumulative effects which may contribute to COVID-19 severity. Therefore, we assumed that antioxidant genetic profile, independently and complemented with laboratory markers, modulates COVID-19 severity. The study included 265 COVID-19 patients. Polymorphism of GSTM1, GSTT1, Nrf2 rs6721961, GSTM3 rs1332018, GPX3 rs8177412, GSTP1 rs1695, GSTO1 rs4925, GSTO2 rs156697, SOD2 rs4880 and GPX1 rs1050450 genes was determined with appropriate PCR-based methods. Inflammation (interleukin-6, CRP, fibrinogen, ferritin) and organ damage (urea, creatinine, transaminases and LDH) markers, complete blood count and coagulation status (d-dimer, fibrinogen) were measured. We found significant association for COVID-19 progression for patients with lymphocytes below 1.0 × 109/L (OR = 2.97, p = 0.002). Increased IL-6 and CRP were also associated with disease progression (OR = 8.52, p = 0.001, and OR = 10.97, p < 0.001, respectively), as well as elevated plasma AST and LDH (OR = 2.25, p = 0.021, and OR = 4.76, p < 0.001, respectively). Of all the examined polymorphisms, we found significant association with the risk of developing severe forms of COVID-19 for GPX3 rs8177412 variant genotype (OR = 2.42, p = 0.032). This finding could be of particular importance in the future, complementing other diagnostic tools for prediction of COVID-19 disease course.


Subject(s)
COVID-19 , Humans , COVID-19/genetics , SARS-CoV-2 , Genotype , Polymorphism, Genetic , Fibrinogen/genetics , Glutathione Peroxidase/genetics , Glutathione Transferase/genetics
5.
Medicina (Kaunas) ; 59(1)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36676788

ABSTRACT

Background and Objectives: In the development of type 2 diabetes mellitus (T2DM) and its complications, genetic and environmental factors play important roles. Diabetic nephropathy (DN), one of the major microangiopathic chronic diabetic complications, is associated with an increased risk of major cardiovascular events and all-cause mortality. The present study was designed to investigate the possible modifying effect of glutathione transferase polymorphisms (GSTM1, GSTT1, GSTP1 rs1138272/rs1695, GSTO1 rs4925 and GSTO2 rs156697) in the susceptibility to T2DM and diabetic nephropathy. Materials and Methods: GSTM1 and GSTT1 deletion polymorphisms were determined by multiplex PCR, whereas GSTO1, GSTO2, and GSTP1 polymorphisms were determined by the real-time PCR in 160 T2DM patients and 248 age- and gender-matched controls. Advanced glycation end products (AGEs) were measured by ELISA. Results: Among six investigated GST polymorphisms, a significant association between the GST genotypes and susceptibility for development of diabetes mellitus was found for the GSTM1, GSTT1, GSTP1 (rs1138272) and GSTO1 polymorphisms. When the GST genotypes' distribution in diabetes patients was assessed in the subgroups with and without diabetic nephropathy, a significant association was found only for the GSTO2 rs156697 polymorphism. Diabetic patients, carriers of the GSTM1 null, GSTT1 null and variant GSTO1*AA genotypes, had significantly increased levels of AGEs in comparison with carriers of the GSTM1 active, GSTT1 active and referent GSTO1*CC genotypes (p < 0.001, p < 0.001, p = 0.004, respectively). Conclusions: The present study supports the hypothesis that GST polymorphisms modulate the risk of diabetes and diabetic nephropathy and influence the AGEs concentration, suggesting the potential regulatory role of these enzymes in redox homeostasis disturbances.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Humans , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Nephropathies/genetics , Genetic Predisposition to Disease , Case-Control Studies , Glutathione Transferase/genetics , Genotype , Glycation End Products, Advanced , Risk Factors
6.
Medicina (Kaunas) ; 58(11)2022 Nov 16.
Article in English | MEDLINE | ID: mdl-36422199

ABSTRACT

Chemotherapy resistance of ovarian cancer, regarded as the most lethal malignant gynecological disease, can be explained by several mechanisms, including increased activity of efflux transporters leading to decreased intracellular drug accumulation, increased efflux of the therapeutic agents from the cell by multidrug-resistance-associated protein (MRP1), enhanced DNA repair, altered apoptotic pathways, silencing of a number of genes, as well as drug inactivation, especially by glutathione transferase P1 (GSTP1). Indeed, GSTP1 has been recognized as the major enzyme responsible for the conversion of drugs most commonly used to treat metastatic ovarian cancer into less effective forms. Furthermore, GSTP1 may even be responsible for chemoresistance of non-GST substrate drugs by mechanisms such as interaction with efflux transporters or different signaling molecules involved in regulation of apoptosis. Recently, microRNAs (miRNAs) have been identified as important gene regulators in ovarian cancer, which are able to target GST-mediated drug metabolism in order to regulate drug resistance. So far, miR-186 and miR-133b have been associated with reduced ovarian cancer drug resistance by silencing the expression of the drug-resistance-related proteins, GSTP1 and MDR1. Unfortunately, sometimes miRNAs might even enhance the drug resistance in ovarian cancer, as shown for miR-130b. Therefore, chemoresistance in ovarian cancer treatment represents a very complex process, but strategies that influence GSTP1 expression in ovarian cancer as a therapeutic target, as well as miRNAs affecting GSTP1 expression, seem to represent promising predictors of chemotherapeutic response in ovarian cancer, while at the same time represent potential targets to overcome chemoresistance in the future.


Subject(s)
MicroRNAs , Ovarian Neoplasms , Humans , Female , Glutathione Transferase , Glutathione S-Transferase pi/genetics , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , MicroRNAs/genetics
7.
PLoS Med ; 17(12): e1003489, 2020 12.
Article in English | MEDLINE | ID: mdl-33301466

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers, with around 9% of patients surviving >5 years. Asymptomatic in its initial stages, PDAC is mostly diagnosed late, when already a locally advanced or metastatic disease, as there are no useful biomarkers for detection in its early stages, when surgery can be curative. We have previously described a promising biomarker panel (LYVE1, REG1A, and TFF1) for earlier detection of PDAC in urine. Here, we aimed to establish the accuracy of an improved panel, including REG1B instead of REG1A, and an algorithm for data interpretation, the PancRISK score, in additional retrospectively collected urine specimens. We also assessed the complementarity of this panel with CA19-9 and explored the daily variation and stability of the biomarkers and their performance in common urinary tract cancers. METHODS AND FINDINGS: Clinical specimens were obtained from multiple centres: Barts Pancreas Tissue Bank, University College London, University of Liverpool, Spanish National Cancer Research Center, Cambridge University Hospital, and University of Belgrade. The biomarker panel was assayed on 590 urine specimens: 183 control samples, 208 benign hepatobiliary disease samples (of which 119 were chronic pancreatitis), and 199 PDAC samples (102 stage I-II and 97 stage III-IV); 50.7% were from female individuals. PDAC samples were collected from patients before treatment. The samples were assayed using commercially available ELISAs. Statistical analyses were performed using non-parametric Kruskal-Wallis tests adjusted for multiple comparisons, and multiple logistic regression. Training and validation datasets for controls and PDAC samples were obtained after random division of the whole available dataset in a 1:1 ratio. The substitution of REG1A with REG1B enhanced the performance of the panel to detect resectable PDAC. In a comparison of controls and PDAC stage I-II samples, the areas under the receiver operating characteristic curve (AUCs) increased from 0.900 (95% CI 0.843-0.957) and 0.926 (95% CI 0.843-1.000) in the training (50% of the dataset) and validation sets, respectively, to 0.936 in both the training (95% CI 0.903-0.969) and the validation (95% CI 0.888-0.984) datasets for the new panel including REG1B. This improved panel showed both sensitivity (SN) and specificity (SP) to be >85%. Plasma CA19-9 enhanced the performance of this panel in discriminating PDAC I-II patients from controls, with AUC = 0.992 (95% CI 0.983-1.000), SN = 0.963 (95% CI 0.913-1.000), and SP = 0.967 (95% CI 0.924-1.000). We demonstrate that the biomarkers do not show significant daily variation, and that they are stable for up to 5 days at room temperature. The main limitation of our study is the low number of stage I-IIA PDAC samples (n = 27) and lack of samples from individuals with hereditary predisposition to PDAC, for which specimens collected from control individuals were used as a proxy. CONCLUSIONS: We have successfully validated our urinary biomarker panel, which was improved by substituting REG1A with REG1B. At a pre-selected cutoff of >80% SN and SP for the affiliated PancRISK score, we demonstrate a clinically applicable risk stratification tool with a binary output for risk of developing PDAC ('elevated' or 'normal'). PancRISK provides a step towards precision surveillance for PDAC patients, which we will test in a prospective clinical study, UroPanc.


Subject(s)
Biomarkers, Tumor/urine , Carcinoma, Pancreatic Ductal/diagnosis , Early Detection of Cancer , Pancreatic Neoplasms/diagnosis , Adult , Aged , Aged, 80 and over , Antigens, Tumor-Associated, Carbohydrate/blood , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/urine , Europe , Female , Humans , Lithostathine/urine , Male , Middle Aged , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/urine , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Trefoil Factor-1/urine , Urinalysis , Vesicular Transport Proteins/urine , Young Adult
8.
Medicina (Kaunas) ; 56(3)2020 Mar 13.
Article in English | MEDLINE | ID: mdl-32183092

ABSTRACT

Background and Objectives: One of the most frequent genetic alterations reported to date in prostate cancer (PC) is aberrant methylation of glutathione transferase P1 (GSTP1). Taking into consideration the involvement of oxidative stress in PC pathogenesis and recent advances in scientific understanding of the role of GSTP1*Ala114Val rs1138272 polymorphism in carcinogenesis, we hypothesized that this single-nucleotide polymorphism (SNP) influences the risk of PC independently of, or in combination with, other GST polymorphisms, including GSTP1*IIe105Val rs1695 or GSTM1 and GSTT1 deletion polymorphisms. Materials and Methods: Genotyping was performed in 237 PC cases and in 236 age-matched controls by multiplex polymerase chain reaction (PCR) for deletion of GST polymorphisms and by quantitative PCR for SNPs. Results: We found that carriers of either GSTP1*Val (rs1138272) or GSTP1*Val (rs1695) variant alleles had a PC risk compared to individuals with both referent alleles (OR = 4.93, 95%CI: 2.89-8.40, p < 0.001 and OR = 1.8, 95%CI: 1.19-2.73, p = 0.006, respectively). Additionally, in a haplotype analysis we found that individuals with GSTP1*C haplotype, represented by both variant alleles (GSTP1*Val rs1695 + GSTP1*Val rs1138272), had a 5.46 times higher risk of PC development compared to individuals with the most frequent haplotype (95%CI = 2.56-11.65, p < 0.001), suggesting a potential role of those variants in PC susceptibility. A regression analysis on the number of risk-associated alleles per individual (GSTM1*active, GSTT1*null, GSTP1*Val rs1695 and GSTP1*Val rs1138272) showed a significant increase in the risk of developing PC, from 3.65-fold in carriers of two risk alleles (95%CI = 1.55-8.61, p = 0.003) to an approximately 12-fold increase in carriers of all four risk alleles (95%CI = 3.05-44.93, p < 0.001). Conclusion: Prostate cancer may be influenced by multiple glutathione transferase (GST) polymorphic genes, especially GSTP1, highlighting the role of gene-gene interactions in human susceptibility to this cancer.


Subject(s)
Glutathione S-Transferase pi/analysis , Polymorphism, Genetic/genetics , Prostatic Neoplasms/genetics , Aged , Case-Control Studies , Genetic Predisposition to Disease , Glutathione S-Transferase pi/blood , Humans , Male , Middle Aged , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/physiopathology , Risk Adjustment/methods , Serbia
9.
Am J Nephrol ; 50(2): 115-125, 2019.
Article in English | MEDLINE | ID: mdl-31238306

ABSTRACT

INTRODUCTION: Overall survival of patients with end-stage renal disease (ESRD) remains poor. Oxidative stress is one of the major risk factors associated with mortality in this patient group. As glutathione S-transferases (GST) are well-established antioxidants, we hypothesized that a model including GST gene polymorphisms, oxidative damage byproducts and cell adhesion markers has a prognostic role in ESRD patient survival. METHODS: A prospective study of 199 patients with ESRD on haemodialysis was conducted. GST genotype, oxidative stress byproducts and cell adhesion molecules were measured in plasma. Multivariate Cox regression and Kaplan-Meier survival analyses were performed to test the predictive ability of these parameters in the 8-year follow-up period. RESULTS: GSTM1-null genotype was associated with significantly shorter overall (HR 1.6, p = 0.018) and cardiovascular-specific (HR 2.1, p = 0.010) survival. Oxidative stress byproducts (advanced oxidation protein products [AOPP], prooxidant-antioxidant balance [PAB], malondialdehyde [MDA]) and cell adhesion molecules (soluble vascular cell adhesion molecule-1 [sVCAM-1] and soluble intercellular adhesion molecule-1 [sICAM-1]) demonstrated a significant predictive role in terms of overall and cardiovascular survival. When 6 biomarkers (GSTM1 genotype, high AOPP/PAB/MDA/-sVCAM-1/sICAM-1) were combined into a scoring model, a significantly shorter overall and cardiovascular survival was observed for patients with the highest score (p < 0.001). CONCLUSION: We identified a novel panel of biomarkers that can be utilized in predicting survival in ESRD patients. This biomarker signature could enable better monitoring of patients and stratification into appropriate treatment groups.


Subject(s)
Cardiovascular Diseases/mortality , Glutathione Transferase/genetics , Kidney Failure, Chronic/mortality , Renal Dialysis , Aged , Biomarkers/analysis , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Clinical Decision-Making , Endothelium, Vascular/metabolism , Endothelium, Vascular/physiopathology , Female , Follow-Up Studies , Humans , Intercellular Adhesion Molecule-1/blood , Intercellular Adhesion Molecule-1/metabolism , Kaplan-Meier Estimate , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/therapy , Male , Malondialdehyde/blood , Malondialdehyde/metabolism , Middle Aged , Oxidative Stress , Patient Selection , Polymorphism, Single Nucleotide , Predictive Value of Tests , Prospective Studies , Risk Assessment/methods , Vascular Cell Adhesion Molecule-1/blood , Vascular Cell Adhesion Molecule-1/metabolism
10.
Medicina (Kaunas) ; 55(8)2019 Aug 03.
Article in English | MEDLINE | ID: mdl-31382611

ABSTRACT

Background: Experimental data show that superoxide dismutase 2 (SOD2) is involved in ochratoxin (OTA)-induced nephrotoxicity, whereas clinical data indicate the role of SOD2 rs4880 or glutathione peroxidase 1 (GPX1) rs1050450 polymorphisms in end-stage renal disease and urothelial carcinoma risk, known to be the major complications of Balkan endemic nephropathy (BEN). Therefore, we hypothesized that SOD2 and GPX1 gene polymorphisms would influence the risk of BEN and its associated tumors. Materials and Methods: The study was conducted in 207 BEN patients and 86 controls from endemic areas. Results: Individuals with both copies of variant SOD2 allele, known for lower mitochondrial antioxidant protection, are at a significantly higher BEN risk (OR = 2.6, p = 0.021). No association was observed between GPX1 gene polymorphism and BEN risk. Combining SOD2 and GPX1 genotypes did not alter the risk of BEN development. Regarding the risk of urothelial tumors in BEN patients, none of the polymorphisms studied was significantly associated with the risk of these tumors. Conclusions: Polymorphism in SOD2 rs4880 gene affects the risk of BEN development. Hence, SOD2 genotyping could, together with a panel of other enzymes, be used as a biomarker of susceptibility in BEN areas.


Subject(s)
Balkan Nephropathy/genetics , Glutathione Peroxidase/genetics , Polymorphism, Genetic/genetics , Superoxide Dismutase/genetics , Aged , Aged, 80 and over , Balkan Nephropathy/epidemiology , Balkan Nephropathy/physiopathology , Biomarkers/analysis , Biomarkers/blood , Bosnia and Herzegovina/epidemiology , Female , Glutathione Peroxidase/blood , Humans , Male , Serbia/epidemiology , Superoxide Dismutase/blood , Glutathione Peroxidase GPX1
11.
Tohoku J Exp Med ; 246(1): 35-44, 2018 09.
Article in English | MEDLINE | ID: mdl-30224590

ABSTRACT

Glutathione S-transferases (GSTs), a superfamily of multifunctional enzymes, play an important role in the onset and progression of renal cell carcinoma (RCC). However, novel GST omega class (GSTO), consisting of GSTO1-1 and GSTO2-2 isoenzymes, has not been studied in RCC yet. Two coding single nucleotide polymorphisms (SNPs) supposedly affect their functions: GSTO1*C419A (rs4925) causing alanine to aspartate substitution (*A140D) and GSTO2*A424G (rs156697) causing asparagine to aspartate substitution (*N142D), and have been associated with several neurodegenerative diseases and cancers. Functional relevance of yet another GSTO2 polymorphism, identified at the 5' untranslated (5'UTR) gene region (GSTO2*A183G, rs2297235), has not been clearly discerned so far. Therefore, we aimed to assess the effect of specific GSTO1 and GSTO2 gene variants, independently and in interaction with established risk factors (smoking, obesity and hypertension) on the risk for the most aggressive RCC subtype, the clear cell RCC (ccRCC). Genotyping was performed in 239 ccRCC patients and 350 matched controls, while plasma levels of 8-hydroxy-2'-deoxyguanosine (8-OHdG), a biomarker of oxidative DNA damage, were determined by ELISA. As a result, combined effect of all three variant genotypes exhibited almost 3-fold risk of RCC development. Additionally, this association was confirmed at the haplotype level [variant GSTO1*A/GSTO2*G (rs156697)/GSTO2*G (rs2297235) haplotype], suggesting a potential role of those variants in propensity to RCC. Regarding the gene-environment interactions, variant GSTO2*G (rs156697) homozygous smokers are at higher ccRCC risk. Association in terms of oxidative DNA damage was found for GSTO2 polymorphism in 5'UTR and 8-OHdG. In conclusion, the concomitance of GSTO polymorphisms may influence ccRCC risk.


Subject(s)
Carcinoma, Renal Cell/genetics , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Kidney Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , 8-Hydroxy-2'-Deoxyguanosine , Case-Control Studies , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Female , Haplotypes/genetics , Humans , Hypertension/genetics , Male , Middle Aged , Obesity/genetics , Risk Factors
12.
Int J Mol Sci ; 19(12)2018 Nov 28.
Article in English | MEDLINE | ID: mdl-30487385

ABSTRACT

Multifunctional enzymes glutathione transferases (GSTs) are involved in the development of chemoresistance, thus representing a promising target for a novel approach in cancer treatment. This superfamily of polymorphic enzymes exhibits extraordinary substrate promiscuity responsible for detoxification of numerous conventional chemotherapeutics, at the same time regulating signaling pathways involved in cell proliferation and apoptosis. In addition to upregulated GST expression, different cancer cell types have a unique GST signature, enabling targeted selectivity for isoenzyme specific inhibitors and pro-drugs. As a result of extensive research, certain GST inhibitors are already tested in clinical trials. Catalytic properties of GST isoenzymes are also exploited in bio-activation of specific pro-drugs, enabling their targeted accumulation in cancer cells with upregulated expression of the appropriate GST isoenzyme. Moreover, the latest approach to increase specificity in treatment of solid tumors is development of GST pro-drugs that are derivatives of conventional anti-cancer drugs. A future perspective is based on the design of new drugs, which would selectively target GST overexpressing cancers more prone to developing chemoresistance, while decreasing side effects in off-target cells.


Subject(s)
Enzyme Inhibitors/therapeutic use , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Animals , Drug Resistance, Neoplasm , Humans , Prodrugs/therapeutic use , Protein Binding
13.
J BUON ; 23(4): 1130-1135, 2018.
Article in English | MEDLINE | ID: mdl-30358222

ABSTRACT

PURPOSE: Purpose: Urothelial bladder cancer (UBC) is the most common malignancy of urinary tract in the developed world. In metastatic UBC, systemic chemotherapy still remains the mainstay of initial treatment. Inter-individual differences in treatment outcome partially may be the consequence of genetic variations in enzymes that modulate oxidative stress. Therefore, we aimed to determine the potential prognostic role of single nucleotide polymorphism (SNP) of the two antioxidant enzymes glutathione peroxidase 1 (GPX1) and superoxide dismutase 2 (SOD2) in metastatic UBC patients treated with cisplatin-based chemotherapy. METHODS: Methods: This prospective single-center hospital-based case-control study included 33 patients with metastatic UBC treated with cisplatin-based chemotherapy and 227 healthy controls. GPX1 SNP (rs1050450) was assessed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP), and SOD2 SNP (rs4880) was determined by quantitative PCR (q-PCR). Overall survival (OS) was evaluated using Kaplan­Meier survival analysis during 2-year follow up period, with the log-rank test for prognostic significance. RESULTS: Results: No significant difference was observed in the distributions of GPX1 and SOD2 gene variants between patients and controls (p˃0.05). Regarding GPX1 polymorphism, no impact of GPX1 polymorphism on OS could be demonstrated (p˃0.05). Finally, Kaplan-Meier survival analysis showed no association between SOD2 polymorphism and OS (p˃0.05). CONCLUSIONS: Conclusions: No association was found between polymorphism of GPX1 and SOD2 and OS in patients with metastatic urothelial bladder cancer treated with cisplatin-based chemotherapy.


Subject(s)
Glutathione Peroxidase/genetics , Superoxide Dismutase/genetics , Urinary Bladder Neoplasms/genetics , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Case-Control Studies , Cisplatin/administration & dosage , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Polymorphism, Single Nucleotide , Prospective Studies , Serbia/epidemiology , Survival Analysis , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/mortality , Glutathione Peroxidase GPX1
14.
Curr Psychiatry Rep ; 19(8): 52, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28681355

ABSTRACT

PURPOSE OF REVIEW: This review aims to provide a brief description of the complex etiology of autism spectrum disorders (ASD), with special emphasis on the recent findings of impaired redox control in ASD, and to suggest a possible model of oxidative stress-specific gene-environment interaction in this group of disorders. RECENT FINDINGS: Recent findings point out to the significance of environmental, prenatal, and perinatal factors in ASD but, at the same time, are in favor of the potentially significant oxidative stress-specific gene-environment interaction in ASD. Available evidence suggests an association between both the identified environmental factors and genetic susceptibility related to the increased risk of ASD and the oxidative stress pathway. There might be a potentially significant specific gene-environment interaction in ASD, which is associated with oxidative stress. Revealing novel susceptibility genes (including those encoding for antioxidant enzymes), or environmental factors that might increase susceptibility to ASD in carriers of a specific genotype, might enable the stratification of individuals more prone to developing ASD and, eventually, the possibility of applying preventive therapeutic actions.


Subject(s)
Autism Spectrum Disorder/metabolism , Gene-Environment Interaction , Oxidative Stress , Prenatal Exposure Delayed Effects/metabolism , Autism Spectrum Disorder/epidemiology , Autism Spectrum Disorder/genetics , Child , DNA Damage , Environmental Exposure/statistics & numerical data , Female , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Humans , Maternal Exposure/statistics & numerical data , Oxidation-Reduction , Pregnancy , Prenatal Exposure Delayed Effects/epidemiology , Prenatal Exposure Delayed Effects/genetics
15.
J BUON ; 22(1): 72-79, 2017.
Article in English | MEDLINE | ID: mdl-28365938

ABSTRACT

PURPOSE: Since several studies have proposed that epithelial ovarian cancer should not be considered as a single disease entity and that it results from an accumulation of genetic changes, we aimed to assess the polymorphic expression of major cytosolic glutathione S-transferases (GSTM1, T1, A1 and P1) with respect to ovarian cancer susceptibility and aggressiveness. METHODS: This case-control study was conducted on 93 newly diagnosed epithelial ovarian cancer patients and 178 healthy matched controls. The multiplex polymerase chain reaction (PCR) was used to detect homozygous deletions of GSTM1 and GSTT1 genes. Analysis of the single nucleotide polymorphism (SNP) GSTA1 C69T was performed using PCR-restriction fragment length polymorphism (RFLP), while for SNP GSTP1 Ile105Val real-time PCR was used. RESULTS: No significant association to ovarian cancer risk was found for individual GSTM1, GSTA1 and GSTP1 genotypes (p>0.05). However, the carriers of GSTT1-active genotype were at 2-fold higher risk of ovarian cancer development (95%CI: 1.00-4.01, p=0.049), which was even more elevated in the subgroup of patients with positive family history of cancer. Moreover, the frequency of all three GST genotypes that might be associated to ovarian cancer risk (GSTT1-active, GSTA1-active and GSTP1-referent) was significantly higher in patients than in the control group (p=0.042). Even more, patients who were carriers of combination of these three genotypes represented over 64% of the total number of patients within any of the International Federation of Gynecology and Obstetrics (FIGO) stages of ovarian cancer. CONCLUSIONS: This study provides supportive evidence that GSTs might affect both susceptibility and progression of ovarian cancer.


Subject(s)
Genetic Predisposition to Disease , Glutathione S-Transferase pi/genetics , Glutathione Transferase/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Polymorphism, Single Nucleotide , Carcinoma, Ovarian Epithelial , Case-Control Studies , Female , Genotype , Humans , Neoplasms, Glandular and Epithelial/enzymology , Neoplasms, Glandular and Epithelial/etiology , Ovarian Neoplasms/enzymology , Ovarian Neoplasms/etiology , Risk
16.
Tohoku J Exp Med ; 240(1): 25-30, 2016 09.
Article in English | MEDLINE | ID: mdl-27568660

ABSTRACT

Balkan endemic nephropathy (BEN) is a chronic familial form of interstitial nephritis that might eventually lead to end stage renal disease. This nephropathy affects individuals living along of the Danube River and its tributaries in Serbia, Bosnia, Croatia, Bulgaria and Romania. The increased incidence of urinary tract tumors in the BEN areas is well described, but its specific genetic predisposition is still unclear. Certain nephrocarcinogenic compounds, including those associated with BEN, are metabolized by glutathione S-transferase (GST) superfamily of phase II detoxication enzymes. Importantly, the GST-mediated detoxification may result in formation of more toxic compounds. We examined the association of common GST polymorphisms and bladder cancer (BC) risk in individuals from BEN areas in Serbia. A hospital-based case-control study included 201 BC cases (67 from BEN region) and 122 controls. Each polymorphism was identified by a PCR-based method. Individuals from BEN region with low-expression GSTA1 genotype (AB+BB) exhibited a 2.6-fold higher BC risk compared to those with GSTA1 (AA) genotype who were from non-BEN region (OR = 2.60, p = 0.015). In contrast, carriers of GSTM1-active genotype from BEN region had a 2.9-fold increased BC risk compared to those with GSTM1-active genotype from non-BEN region (OR = 2.90, p = 0.010). Likewise, carriers with GSTT1-active genotype from BEN region exhibited 2.1-fold higher BC risk compared to those from non-BEN region with GSTT1-active genotype (OR = 2.10, p = 0.027). Thus, common polymorphisms in GSTA1, GSTM1 and GSTT1 are associated with susceptibility to BC in individuals from BEN areas of Serbia.


Subject(s)
Balkan Nephropathy/genetics , Genetic Predisposition to Disease , Glutathione Transferase/genetics , Polymorphism, Genetic , Urinary Bladder Neoplasms/enzymology , Urinary Bladder Neoplasms/genetics , Case-Control Studies , Demography , Female , Humans , Male , Middle Aged , Risk Factors , Serbia
17.
BMC Nephrol ; 15: 12, 2014 Jan 14.
Article in English | MEDLINE | ID: mdl-24423050

ABSTRACT

BACKGROUND: The presence of glutathione transferase (GST) M1 null genotype (GSTM1-null) in end-stage renal disease (ESRD) patients is associated with lower overall survival rate in comparison to those with GSTM1-active variants. We examined association between GSTM1 and GSTT1 deletion polymorphisms as well as SNPs in GSTA1/rs3957357 and GSTP1/rs1695 genes with overall and cause-specific cardiovascular mortality in ESRD patients. METHODS: Total of 199 patients undergoing hemodialysis were included in the study. Median value of time elapsed from dialysis initiation until the death, or the end of follow-up was 8 ± 5 years. The effect of GSTM1, GSTT1, GSTP1 and GSTA1 gene polymorphisms on predicting overall and specific cardiovascular outcomes (myocardial infarction, MI or stroke) was analyzed using Cox regression model, and differences in survival were determined by Kaplan-Meier. RESULTS: GSTM1-null genotype in ESRD patients was found to be independent predictor of overall and cardiovascular mortality. However, after false discovery rate and Bonferroni corrections this effect was lost. The borderline effect modification by wild-type GSTA1*A/*A genotype on associations between GSTM1-null and analyzed outcomes was found only for death from stroke. Homozygous carriers of combined GSTM1*0/GSTA1*A genotype exhibited significantly shorter time to death of stroke or MI in comparison with carriers of either GSTM1-active or at least one GSTA1*B gene variant. The best survival rate regarding cardiovascular outcome was found for ESRD patients with combined GSTM1-active and mutant GSTA1*B/*B genotype. CONCLUSIONS: Combined GSTM1*0/GSTA1*A genotypes might be considered as genetic markers for cardiovascular death risk in ESRD patients, which may permit targeting of preventive and early intervention.


Subject(s)
Cardiovascular Diseases/mortality , Death, Sudden, Cardiac/epidemiology , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Glutathione Transferase/genetics , Renal Dialysis/mortality , Female , Genetic Association Studies , Genetic Markers/genetics , Humans , Incidence , Male , Middle Aged , Risk Factors , Serbia/epidemiology , Survival Rate
18.
Clin Exp Ophthalmol ; 42(3): 277-83, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23927022

ABSTRACT

BACKGROUND: Glutathione S-transferase omega-1 and 2 have a unique range of enzymatic activities, including the regeneration of ascorbate by their dehydroascorbate reductase activities. Because these enzymes could have a protective role from oxidative damage in the lens, the question of whether the two coding glutathione S-transferase omega polymorphisms confer the risk of age-related cataract was addressed. METHODS: rs4925 (Ala140Asp) of glutathione S-transferase omega-1 and rs156697 (Asn142Asp) of glutathione S-transferase omega-2 polymorphisms in 100 patients with age-related cataract and 130 controls were assessed. RESULTS: Presence of one mutant GSTO1*Asp or GSTO2*Asp allele did not contribute independently towards the risk of cataract; however, homozygous carriers of GSTO1*Asp/GSTO2*Asp haplotype demonstrated 3.42-fold enhanced risk of cataract development (95% confidence interval = 0.84-13.93; P = 0.086). When GSTO genotype was analysed in association with smoking or professional exposure to ultraviolet irradiation, carriers of at least one mutant GSTO2*Asp allele had increased risk of cataract development in comparison with individuals with wild-type GSTO2*Asn/Asn with no history of smoking or ultraviolet exposure (odds ratio = 6.89, 95% confidence interval = 1.81-16.21, P = 0.005; odds ratio = 4.10, 95% confidence interval = 1.23-13.74, P = 0.022, respectively). Regarding the distribution of particular glutathione S-transferase omega genotype and cataract type, the highest frequency of mutant GSTO2*Asp allele was found in patients with nuclear cataract. CONCLUSION: The results indicate that mutant GSTO2*Asp genotype is associated with increased risk of age-related cataract in smokers and ultraviolet-exposed subjects, suggesting a role of inefficient ascorbate regeneration in cataract development.


Subject(s)
Cataract/genetics , Glutathione Transferase/genetics , Lens, Crystalline/radiation effects , Polymorphism, Single Nucleotide , Radiation Injuries/genetics , Smoking/genetics , Ultraviolet Rays/adverse effects , Aged , Aging , Alleles , Female , Genetic Predisposition to Disease , Genotype , Genotyping Techniques , Humans , Male , Polymerase Chain Reaction , Polymorphism, Restriction Fragment Length , Risk Factors
19.
Nephrol Dial Transplant ; 28(1): 202-12, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23034843

ABSTRACT

BACKGROUND: Increased oxidative stress is a hallmark of end-stage renal disease (ESRD). Glutathione S-transferases (GST) are involved in the detoxification of xenobiotics and protection of oxidative damage. We hypothesized that genetic polymorphism in antioxidant enzymes GSTA1, GSTM1, GSTP1 and GSTT1 is more frequent in ESRD and modulates the degree of oxidative stress in these patients. METHODS: GSTA1, GSTM1, GSTP1 and GSTT1 genotypes were determined in 199 ESRD patients and 199 age- and gender-matched controls. Markers of protein and lipid oxidative damage [thiol groups, carbonyl groups, advanced oxidative protein products, nitrotyrosine, malondialdehyde (MDA) and MDA adducts], together with total oxidant status and pro-oxidant-antioxidant balance were determined. RESULTS: Individual GST polymorphisms influence vulnerability to both protein and lipid oxidation, with GSTM1-null gene variant having the most pronounced effect. Furthermore, a strong combined effect of null/low-activity GSTM1, GSTT1, GSTA1 and GSTP1 genotypes in terms of susceptibility towards oxidative and carbonyl stress was found in ESRD patients. When patients were stratified according to GSTM1 and GSTT1, the highest oxidant damage was noted in those with the GSTM1-null/GSTT1-null genotype. The observed effect was even stronger in patients with the third low-activity GSTP1 or GSTA1 genotype. Finally, the level of oxidative and carbonyl stress was most pronounced in the subgroup of patients with all four null or low-activity GSTM1, GSTT1, GSTP1 and GSTA1 genotypes. CONCLUSIONS: According to the GST genotype, ESRD patients may be stratified in terms of the level of oxidative and carbonyl stress that might influence cardiovascular prognosis, but could also improve efforts towards individualization of antioxidant treatment.


Subject(s)
Glutathione Transferase/genetics , Kidney Failure, Chronic/genetics , Oxidative Stress/genetics , Renal Dialysis/adverse effects , Aged , Biomarkers , Female , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Polymorphism, Genetic
20.
J Clin Lab Anal ; 27(2): 105-12, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23349048

ABSTRACT

BACKGROUND: To assess the prognostic significance of four inflammatory markers (TNF-α, high sensitive C-reactive protein (hs-CRP), intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1)) in chronic heart failure (CHF) patients with respect to individual outcomes, especially disease exacerbation and mortality. METHODS: Plasma adhesion molecules, ICAM-1, and VCAM-1, together with TNF-α and hs-CRP were determined in 120 CHF patients and 69 healthy controls. Endothelial function was also estimated by flow-mediated brachial artery dilatation. RESULTS: Increased levels of all investigated inflammatory markers were found in CHF patients compared to controls, with the rise more pronounced in New York Heart association (NYHA) functional IV class. Significant correlations were obtained for VCAM-1 and brain natriuretic peptide (r = 0.191; P = 0.038), as well as, ICAM-1 and endothelium-dependent vasodilatation (r = -0.235; P = 0.01). Kaplan-Meier analysis showed disease exacerbation in patients with TNF-α levels >2.78 pg/ml significantly shorter compared to those with TNF-α levels <2.78 pg/ml (log-rank test = 8.270; P = 0.004), while similar association was observed for patients with hs-CRP levels >4.76 mg/l (log-rank test = 5.052; P = 0.025) and VCAM-1 levels >1200 ng/l (log-rank test = 5.45; P = 0.020) with respect to mortality. Cox regression analysis demonstrated only VCAM-1 (HR = 4.7; 95% confidence interval (CI): 1.1-18.7; P = 0.030) as independent death predictor, while TNF-α was associated with disease exacerbation (HR = 8.2; 95%CI: 1.1-23.0; P = 0.045). CONCLUSIONS: VCAM-1 appears to be useful in risk stratification of CHF patients and in screening, to identify subjects at risk for heart failure related events.


Subject(s)
Heart Failure/blood , Tumor Necrosis Factor-alpha/blood , Vascular Cell Adhesion Molecule-1/blood , Aged , Analysis of Variance , Biomarkers/blood , C-Reactive Protein/metabolism , Chronic Disease , Female , Humans , Inflammation/blood , Intercellular Adhesion Molecule-1/blood , Kaplan-Meier Estimate , Male , Middle Aged , Morbidity
SELECTION OF CITATIONS
SEARCH DETAIL