Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Jun 27, 2021; 13(6): 620-633
Published online Jun 27, 2021. doi: 10.4254/wjh.v13.i6.620
Glutathione-S-transferases genes-promising predictors of hepatic dysfunction
Vasyl Prysyazhnyuk, Kateryna Bobkovych, Department of Propedeutics of Internal Diseases, Bukovinian State Medical University, Chernivtsi 58002, Chernivtsi region, Ukraine
Larysa Sydorchuk, Valentina Dzuryak, Department of Family Medicine, Bukovinian State Medical University, Chernivtsi 58002, Chernivtsi region, Ukraine
Ruslan Sydorchuk, Department of Surgery, Bukovinian State Medical University, Chernivtsi 58002, Chernivtsi region, Ukraine
Iryna Prysiazhniuk, Inna Buzdugan, Department of Internal Medicine and Invectious Diseases, Bukovinian State Medical University, Chernivtsi 58002, Chernivtsi region, Ukraine
Petro Prysyazhnyuk, Department of Medical and Pharmaceutical Chemistry, Bukovinian State Medical University, Chernivtsi 58002, Chernivtsi region, Ukraine
ORCID number: Vasyl Prysyazhnyuk (0000-0001-8932-9512); Larysa Sydorchuk (0000-0001-9279-9531); Ruslan Sydorchuk (0000-0002-3603-3432); Iryna Prysiazhniuk (0000-0002-9441-5498); Kateryna Bobkovych (0000-0001-5783-2412); Inna Buzdugan (0000-0003-0425-8845); Valentina Dzuryak (0000-0002-8810-3164); Petro Prysyazhnyuk (0000-0003-3426-4365).
Author contributions: Prysyazhnyuk V was responsible for conception and design, analysis and interpretation of the clinical data, critical revision of the manuscript, final approval of the article; Sydorchuk L, Sydorchuk R analysed and interpreted of the pathophysiological data, critical revision of the manuscript, final approval of the article; Prysiazhniuk I was responsible for acquisition of data, drafting of the article, critical revision of the manuscript, final approval of the article; Bobkovych K, Buzdugan I, Dzhuryak V, and Prysyazhnyuk P were responsible for acquisition of data, critical revision of the manuscript, final approval of the article.
Conflict-of-interest statement: The authors declare no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Vasyl Prysyazhnyuk, DSc, MD, PhD, Professor of the Department of Propedeutics of Internal Diseases, Bukovinian State Medical University, Teatralna square 2, Chernivtsi 58002, Chernivtsi region, Ukraine. prysyaznyuk_v@ukr.net
Received: March 17, 2021
Peer-review started: March 17, 2021
First decision: May 2, 2021
Revised: May 6, 2021
Accepted: June 3, 2021
Article in press: June 3, 2021
Published online: June 27, 2021

Abstract

One of the most commonly known genes involved in chronic diffuse liver diseases pathogenesis are genes that encodes the synthesis of glutathione-S-transferase (GST), known as the second phase enzyme detoxification system that protects against endogenous oxidative stress and exogenous toxins, through catalisation of glutathione sulfuric groups conjugation and decontamination of lipid and deoxyribonucleic acid oxidation products. The group of GST enzymes consists of cytosolic, mitochondrial and microsomal fractions. Recently, eight classes of soluble cytoplasmic isoforms of GST enzymes are widely known: α-, ζ-, θ-, κ-, μ-, π-, σ-, and ω-. The GSTs gene family in the Human Gene Nomenclature Committee, online database recorded over 20 functional genes. The level of GSTs expression is considered to be a crucial factor in determining the sensitivity of cells to a broad spectrum of toxins. Nevertheless, human GSTs genes have multiple and frequent polymorphisms that include the complete absence of the GSTM1 or the GSTT1 gene. Current review supports the position that genetic polymorphism of GST genes is involved in the pathogenesis of various liver diseases, particularly non-alcoholic fatty liver disease, hepatitis and liver cirrhosis of different etiology and hepatocellular carcinoma. Certain GST allelic variants were proven to be associated with susceptibility to hepatological pathology, and correlations with the natural course of the diseases were subsequently postulated.

Key Words: Glutathione-S-transferase, Non-alcoholic fatty liver disease, Drug induced liver disease, Liver cirrhosis

Core Tip: Current review provide data regarding impact of genetic polymorphism of glutathione-S-transferase (GST) genes in the pathogenesis of various liver diseases, particularly non-alcoholic fatty liver disease, hepatitis and liver cirrhosis of different etiology and hepatocellular carcinoma. Certain GST allelic variants were proven to be associated with susceptibility to hepatological pathology and correlations with the natural course of the diseases were subsequently postulated.



INTRODUCTION

Glutathione-S-transferases (GSTs) are group of phase II detoxification enzymes that catalyses the conjugation of glutathione (GSH) to a variety of endogenous and exogenous electrophilic compounds. It is without doubts that phase I enzyme reaction catalyses the incorporation of a functional group to a foreign compound, resulting in the formation of an intermediate metabolite. However, many of intermediates contain high potent chemical groups that can react with different cellular components including DNA, proteins and lipids[1,2]. This presence of intermediate metabolites can lead to multiple adverse health effects. Intermediate substances undergo phase II metabolism to form highly hydrophilic and less chemically active compounds, facilitating their excretion through bile or urine. Moreover, before being eliminated from the body, an extraneous compound can directly take part in phase II bypassing phase I detoxification. Phase II enzymes deactivate and detoxify foreign compounds unlike phase I enzymes which serves as activation metabolism, and therefore referred to as detoxification enzymes[3-5]. The aim of the current review was to overview up-to-date data and sum up results of own investigations regarding the distribution of GST genes polymorphisms, possible mechanisms of their involvement in the processes of desintoxication, drugs metabolism and cancerogenesis, and their role in the natural course of various liver diseases.

GSTs are presented by the cytosolic and membrane-bound microsomal super-family members. The groups of microsomal GSTs are structurally distinct from the cytosolic enzymes as they are rather homo-and heterotrimerise than dimerise in order to form a solitary active site. Microsomal GSTs are known to be the primary players in the endogenous metabolism of certain important substances like prostaglandins and leukotrienes. In contradistinction to microsomal GSTs, cytosolic GSTs are highly polymorphic and can easily be divided into eight sub-classes: α, μ, ω, π, θ, ζ, σ-, and ω-. The π and μ classes of GSTs play a regulatory role in the mitogen-activated protein kinase pathway participating in cellular survival and death signaling via protein-protein interactions with c-Jun N-terminal kinase 1 (JNK1) and apoptosis signal-regulating kinase (ASK1). JNK and ASK1 are in turn activated in response to cellular stress[6-8].

GSTs are broadly distributed in the living world, from single cell organisms like bacteria to various plants, animals, and humans. Plant GSTs include the φ, τ, θ, ξ and λ classes; the θ and ξ have analogues in animals, too. Moreover, the ξ and θ classes are numerous in non-vertebrate animals. Advocating that the ancestral progenitor for mammalian GSTs, probably arose from the θ class GSTs based on significant homology between the θ class GST and a dichloromethane dehalogenase enzyme from the prokaryote methylobacteriaceae, belonging to the genus of rhizobiales which is known to be able to undergo genetic transformation and become competent for DNA uptake close to the end of the exponential growth phase[9-12].

The review of the GSTs gene family in the Human Gene Nomenclature Committee (HGNC), online database, shows 23 (as for beginning of 2021) functional genes contained within the group[13], which is a minor upgrade from the last decade, when there were only 21 of such genes reported. However, the number of subfamilies varies from 16 to 26 in different sources, and some genes of the group were determined as encoding membrane-bound enzymes having GST-like activity, but these genes are not related to the GSTs gene family evolutionarily. These genes include GST-κ1 [glutathione S-transferase kappa 1 (GSTK1), GST13, HGNC: 16906, Chromosome 7q34], and microsomal glutathione S-transferase 1 (MGST1, Chromosome12p12.3) microsomal glutathione S-transferase 1-like 1 (prostaglandin E synthase-PTGES, MGST-IV, PIG12, MGST1-L1, TP53I12, HGNC: 9599, Chromosome 9q34.11), microsomal glutathione S-transferase 2 (MGST2, MGST-II, HGNC: 7063, Chromosome 4q31.1), and microsomal glutathione S-transferase 3 (MGST3, GST-III, HGNC: 7064, Chromosome 1q24.1). The known human GSTs gene family consists of six subfamilies-α (GSTA-alpha), μ (GSTM-miu), ω (GSTO-omega), π (GSTP-pi), θ (GSTT-theta) and ξ (GSTZ-zeta)[14].

Probably, naming of GSTs genes can cause confusion, because both GSTW and GSTO names are similarly used for GST omega (ω) subfamily marking, and GSTT or GSTQ are concurrently used for GST theta (τ) subfamily listing in different sources. The reason for this lack of certainty originates from the HGNC’s rules. Moreover, quite similar nomenclature problems were reported with the mouse GST genes[14,15].

Nonetheless, while only human GSTs are of valid clinical significance, other GSTs genes are of notable interest as this may explain both the connections and developments of human GSTs. The soluble GSTs can be subdivided into the cytosolic and mitochondrial forms, only GSTκ is exclusively mitochondrial, while GTSA1, 4, GSTM1 and GSTP1 encode both cytosolic and mitochondrial forms. The rest of the GSTs genes encode cytosolic proteins only. Note worthily, a vast number of GSTs were first identified in non-mammalian organisms, and were later recognised in humans and mammals[16-18], however most of the mammalian GSTs have been extensively studied and classified according to commonly assented criteria.

NON-HUMAN GSTS

Reports concerning plant GST enzyme revealed its involvement in catalysing the detoxification of the herbicide atrazine by conjugation to the endogenous γ-L-glutamyl-L-cysteinyl-glycine in sorghum and maize plants, which initiated a research that focuses on the detoxification of various herbicides and other toxic xenobiotic compounds in plants[19]. GSTs exhibit catalysis of the conjugation between various xenobiotics with electrophilic centres and the nucleophilic GSH, tagging the xenobiotic for vacuolar sequestration. The resulting γ-L-glutamyl-L-cysteinyl-β-alanine conjugates were much less toxic and more water-soluble than the original xenobiotics. It was shown that multiple plant GSTs participate in antioxidative protection due to their glutathione peroxidase activity[20].

The floral GSTs are mostly cytosolic and can represent up to 2% of soluble proteins. They have the ability to manifest auxin-inducibility and have ligandin function as well to participate in auxin transport. GSTs play a significant role during the normal metabolism of plant secondary products like anthocyanins[21]. The understanding of GSTs' role in endogenous floral processes and metabolic substrates had been still far from complete in contrast to the vast knowledge collected about their detoxification function[20,22].

Likewise, in human genome, floral GSTs enzymes are encoded by large gene families. The genome of the model plant Arabidopsis thaliana harbors 54 GST genes, which are grouped into seven distinct classes in plants. The well-studied large GSTF and GSTU classes are specific to plants, whilst the smaller GSTZ and GSTT classes exist in animal and human tissues. Lesser data is obtainable about the three outlying minor classes including GSTL, dehydroascorbate reductases, and tetrachlorohydroquinone dehalogenase[21,23].

HUMAN GSTS

Human GSTs genes have multiple and frequent polymorphisms, including the complete absence (up to 20%-50% in some groups and populations) of the GSTM1 or the GSTT1 gene. The prevalence of the null genotype of GSTT1 and GSTM1 genes are heterogeneous amongst different ethnic populations. The GSTT1 deletion is found in 20% of Caucasians and 80% of Asians[24]. While GSTM1 zero genotype is detected in 38%-67% of Caucasian individuals, 33%-63% in East Asians and 22% to 35% in Africans and African Americans[25]. The substitution of adenine for guanine in nucleotide position 313 in the GSTP1 gene leads to a reduction in the GST enzymatic activity which plays a significant role in the development of various diseases[26].

Following deficit in evident GSTs activities may lead to impaired detoxication of environmental substances, like toxins, carcinogens or drugs that may consequently generate clinically worth problems in patients lacking these genes[14,27-29].

GSTA, GSTM, and GSTP are over expressed in rat model of hepatic neoplasms (preneoplastic nodules) and the increased levels of these isoenzymes are assumed to provide the multidrug-resistant phenotype observed in these lesions. The majority of human tumors and human tumor cell lines express significant amounts of GSTP. The mechanisms responsible for over expression of GSTs, implicate transcriptional activation, stabilization of either messenger ribonucleic acid or protein, and gene amplification. In humans, remarkable interindividual differences are present in the expression of GSTA, GSTM, and GSTT. However, the exact molecular basis for the variation in GSTA is not known; missing of certain GSTM and GSTT classes can be attributed to deletion of the GSTMI gene in 50% of the population and deletion of the GSTTI gene in 16% of the population. The biological consequences of failure to express hGSTMI or hGSTTI protein can include higher susceptibility to some types of malignancies including skin, colon, bladder, and possibly lung cancer[10,30].

The level of GSTs expression is considered to be a crucial factor in determining the sensitivity of cells to a broad spectrum of toxins. The most abundant mammalian GSTs are the GSTA, GSTM and GSTP, however the biological control of these families is complex as they exhibit species-, age-, sex-, tissue-, and tumor-specific patterns of expression. Moreover, GSTs as shown above are regulated up and down by a broad spectrum of xenobiotics and drugs, with a significant number of these substances occurring naturally as non-nutritional components in modern food. It is obvious that humans are exposed regularly to such compounds[10].

Majority of chemical compounds, acting as GSTs inducers or inhibitors, have effect on transcriptional activation of GSTs genes through either antioxidant-responsive element, xenobiotic-responsive element, GSTP enhancer l, or glucocorticoid-responsive element[31,32].

The probability of GSTs is regulated in vivo by reactive oxygen species which is based on evidence that is not only but some of the most potent. GSTs inducers are capable of generating free radicals by redox-cycling, but hydrogen peroxide has been shown to strongly induce GSTs in plant and mammalian cells. An induction of GST by reactive oxygen species would appear to represent an adaptive response as GSTs detoxify some of the toxic peroxide-, carbonyl-, and epoxide-containing metabolites produced within the cell during oxidative stress[33-35].

Several functional studies of individual GSTs showed that they can positively contribute to host resistance against various microorganisms, whereas some physiologic mechanisms undergo further studying. Notwithstanding, the elevated total GST enzyme activities and notable accumulation of multiple GST transcripts and proteins was often observed in numerous host-pathogen interactions[23,36]. GSH is the most important non-protein thiol compound in several organisms and plays an important role in signaling and host defense reactions in infection. GSTs' participation in antioxidative react together with the crucial cellular antioxidant GSH in order to eliminate lipid hydroperoxides that accumulate in infected tissues, is clearly their distinguishable function[37-39].

Substantiation of GSTs genes from some commensals and parasites that may have immunomodulatory effect towards the immune system is growing, based on the involvement of separate profiles of cytokine gene transcription and different patterns of cell growth. Both antioxidants and oxidative stress manifest prompt transcription effect on many of the GSTs genes, which leads to increased protection of the cell against insult caused by environmental chemicals and drugs[40-42].

Possible interactions between host and microorganisms may result in three different ways: resistance gene (R-gene) mediated resistance, basal resistance and virulence. The first one (R-gene mediated), hypersensitive-type resistance is based on a specific interaction of a bacterial effect or gene product with the R-gene of the host organism. R-gene mediated type of resistance is commonly corresponded with the localised cell death in infected host. It is unspecific, in case of basal resistance recognition; opposite to the R-gene mediated cell death, as genetically alien organisms are recognised based on their common molecular patterns. Induction of basal resistance is not associated with perceptible symptoms, in contrast to the hypersensitive-type R-gene mediated cell death. Poor host defense results in virulence[32,43].

Several members of the cytosolic GSTA, GSTM, GSTP, GSTT, microsomal transferases MGST2 and MGST3, are up-regulated by a wide spectrum of foreign compounds including but not limited to fumaric acid, thiazolidinediones, dexamethasone, phenobarbital, β-naphthoflavone, oltipraz, sulforaphane, coumarin, etc.[42]. The mechanism explaining this gene expression induction includes the aryl hydrocarbon receptor, and rostane receptor, the Pregnane X receptor, nuclear factor erythroid 2-related factor 2, CAATT/enhancer binding protein-β, and peroxisome proliferator-activated receptor-γ, which connects GSTs with other pathogenetic mechanisms, genes, and clinical conditions that include insulin resistance, diabetes mellitus type 2, arterial hypertension and abdominal obesity[44].

Due to the fact that GSTs play a determinative role in the detoxification of xenobiotics, their down- or up-regulation may obviously affect biological effects and metabolism of many biologically active compounds, industrials and environmental pollutants. Several studies have demonstrated the potency of some flavonoids to modify the expression of GSTs and their activities. Furthermore, real effect of flavonoid compounds on GSTs strongly hinge on concentration, remedy administration duration, chemical structure of particular flavonoid, as well as on GST origin and isoform. To add confusion, in vitro and in vivo studies results are often inconsistent, incongruous or conflicting. Notwithstanding, prudential use of a flavonoid enriched diets, which may potentially induce GSTs are commonly beneficial, however the uncontrolled intake of certain flavonoids like catechins and quercetin in high doses as a dietary supplement may threaten health in consequence of GST inhibition. Moreover, combined use of certain flavonoids with drugs (acetaminophen, cisplatine, cyclophosphamide, and simvastatin) or xenobiotics (acrylamide, isocyanates polycyclic aromatic hydrocarbons, and chlorpyrifos), which are GSTs substrates, might have significant pharmacological and toxicological consequences[45].

GSTs genes often, demonstrate high inductivity through various stimuli of both abiotic and biotic origin. For example, salicylic acid (SA) showed prompt inducible effect on multiple GSTs. Some of the GSTs genes (GSTF2, GSTF8, GSTF10, GSTF11) are recognised determining SA-binding receptor proteins, though the biological relevance of SA binding to these GSTs needs further study[36,46-48].

Similar behavior may be observed in other genes involving in hepato-pancreatic conditions like angiotensin-converting enzyme gene and peroxisome proliferator-activated receptors-γ gene[49]. We can presume, that there is little evidence of specific precise cellular hepatic alteration mechanisms resulted from GST enzymes dysfunction or corresponding genetics' dysregulations.

NONALCOHOLIC FATTY LIVER DISEASE

Due to the studies of possible difference in the distribution frequency of allelic variations in the GSTP1 A313G polymorphism, it has been established that G allele is spread significantly and more frequent in patients with nonalcoholic fatty liver disease (NAFLD) than in healthy individuals (χ2 = 5.69, P = 0.017) in Ukrainian population (Table 1)[50]. This data is consonant with the results of Hashemi et al[51], who have demonstrated that G allele of GSTP1 gene is a risk factor for NAFLD formation. It was investigated, that total bilirubin level in blood of NAFLD patients with GG genotype of A313G polymorphism of GSTP1 gene was higher as compared to AA genotype and AG genotype carriers. Presence of G allele was also associated with increased alaninaminotransferase activity, which was noticed to be significantly higher in NAFLD patients AG, and GG genotypes carriers as compared to patients with AA genotype[52].

Table 1 Distribution of polymorphic variants of the A313G polymorphism of the GSTP1 gene in patients with nonalcoholic fatty liver disease and healthy individuals.
Genotypes of the gene GSTP1
Patients with NAFLD, n = 104
Healthy individuals, n = 45
Absolute number, n
%
Absolute number, n
%
AA4745, 2%2862, 2%
AG 4240, 4%1635, 6%
GG1514, 4%12, 2%
A-allele13665, 4%7280, 0%
G-allele7234, 6%1820, 0%

Pro-and anti-inflammatory cytokines and adipokines profile varies in NAFLD patients with different polymorphic variants of the GSTP1 gene (A313G) in particular. Homozygous patients with G allele are characterised by higher level of interleukin-10 (I1-10) in the blood as compared to patients with the AA and AG genotypes, that may occur potentially in response to the increase in the tumor necrosis factor-α (TNF-α) concentration, which proved the increased activity of inflammation processes[53,54]. NAFLD patients were investigated with low adiponectin levels in the blood in comparison with healthy people[55]. Moreover, according to Li et al[56] low adiponectin level is associated with the progression of steatohepatitis. The adiponectin concentration was lower in patients with NAFLD and AG and GG genotypes than in those with the AA genotype, indicating a worse adipokine profile for the NAFLD natural course[50]. A reverse tendency has been determined for leptin, however its blood level was higher in NAFLD patients with AG and GG genotypes as compared to those with the AA genotype[50]. This elevation of the leptin content in the GSTP1 G allele carriers was, probably, associated with a high TNF-α concentration stimulating leptin production[57]. The aforementioned can prove the development of the leptin-resistance syndrome more severe in this cohort of patients[58]. In general, these observations indicate the formation of adipokine imbalance in the examined patients with AA genotype, which is typical for patients with NAFLD[59] which causes elevated leptin concentration against decrease adiponectin level in the blood[60].

Deletion polymorphic variants of GSTT1 and GSTM1 genes prevalence amongst NAFLD patients was approximately the same as their distribution between healthy individuals in Ukrainian population. These data are partially different from those suggested by Hori et al[61] who reported higher frequency of GSTM1 null genotype in NAFLD patients as compared to control in the Japanese. There were not any notable differences in the parameters of the synthetic, detoxification, excretory liver functions together with activity of cytolytic and cholestatic syndromes and lipid profile in NAFLD patients with deletion of GSTT1 and GSTM1 genes and patients with functional allele of these genes[62]. It agrees with Rafiee et al[63] who also did not define importance contrasts in cholesterol and triglycerides plasma levels in individuals with different polymorphic variants of the studied genes. Interestingly, earlier studies of Maciel et al[64] suggested that double deletion genotypes of GSTM1 and GSTT1 genes were associated with hypertriglyceridemia.

Elevated TNF-α level in the blood is typical for NAFLD patients as compared to healthy individuals[65]. Jamali et al[66] proposed an algorithm involving TNF-α for predicting NAFLD/non-alcoholic steatohepatitis. Importantly, that null-genotype of GSTT1 gene goes with higher TNF-α concentration as compared with patients having allele variant of GSTT1, and thereby indicate the activation of proinflammatory segment of cytokine profile and inflammatory processes[62]. Note worthily, TNF-α is one of the key factors involved in the insulin resistance, inflammation and apoptosis in case of NAFLD[67], thus its elevated level could be a predictor of aggravated liver injury in NAFLD patients with null-genotype of GSTT1 gene.

Certain peculiarities in adipokine profile were detected regarding GSTM1 genotype. Leptin plasma level was significantly higher in patients with null-genotype of GSTM1 gene as compared to NAFLD patients with functional allele. This elevation of leptin content in null-genotype GSTM1 carriers was probably associated with a high TNF-α concentration that stimulates leptin production[57]. Deletion polymorphism of GSTT1 and GSTM1 genes in patients with NAFLD was associated with lower content of restored glutathione, catalase activity. And in the case of carrier of zero genotype of GSTM1 gene; it was also with higher level of reaction products of thiobarbituric acid in blood as compared to patients with functional allele of the gene[68].

DRUG INDUCED LIVER INJURY AND HEPATITIS

Prevalence of G allele of GSTP1 (A313G) gene did not differ notably in chronic hepatitis patients in comparison with healthy individuals in Ukrainian population, however, presence of G allele was associated with higher activity of cytolytic syndrome lower restored glutathione blood content in comparison with patients AA genotype carriers[69]. GSTP1 IIe/Val genotype was significantly more frequent in the patients with chronic hepatitis B infection and in patients with cirrhosis than in healthy individuals in Turkey; GSTP1 Val/Val genotype was even more frequent in these patients[70]. In addition, these authors denoted relation between GSTP1 gene polymorphism and hepatitis stage. In fact, as IIe/Val and Val/Val genotype frequencies increased so did the stages of the disease and tendency grow towards cirrhosis[70].

In our previous study, it was found that deletion genotype of GSTM1 and GSTT1 in patients with chronic hepatitis were representative to those in healthy individuals. Qi et al[71], have discovered that the genes GSTM3 and GSTP1 promoter methylation, which causes dysfunction of intracellular antioxidant defense system, more frequently occurs in patients with acute and chronic liver failure in case of hepatitis B virus, compared to patients with compensated viral hepatitis. Determination of methylated promoters of GSTP1 and GSTM3 genes can serve as a prognostic factor in the development of acute and chronic liver failure in these patients. It was found that GSTO2 mutant genotypes were increased with progression, and the degree of hepatitis B virus (HBV) infection and the patients had mutant GSTO2 genotypes such as (A/G, and G/G) were more susceptible for more severe HBV disease progression. The authors of the aforementioned study concluded that people with A/G and G/G genotype for GSTO2 are more prone to develop hepatic failure[72]. Certain investigations have driven to the relation of GST gene polymorphism and drug induced liver injury. It was discovered almost twenty years ago, that homozygous null mutation at the GSTM1 gene might predispose to hepatotoxicity for drugs used for the treatment of tuberculosis[73]. This statement was supported in the following studies reviling GSTT1 homozygous null polymorphism may be a risk factor of antituberculosis drug-induced hepatotoxicity in Caucasians[74]. Meanwhile, presence of at least one functional allele of GSTM1 was significantly more frequent amongst the groups with higher grades of liver toxicity for antituberculosis drugs in Brazilians[75]. Contrarily, GSTT1 and GSTM1 were not related to increased antituberculosis drug induced liver injury in Indian citizens[76]. By now, certain researchers[77] have linked troglitazone intoxication in the development of chronic diffuse liver diseases with the double-zero genotype GSTT1 and GSTM1 genes, considering its consequence of insufficient activity of detoxification defense systems, low activity of conjugation of sulfuryl groups. It has been shown that the zero genotype of GSTT1 gene increases the risk of drug-induced liver damage in particular, due to the use of isoniazid[78]. Finally, in meta-analysis, it was found that null GSTM1 genotype was responsible for higher susceptibility to drug induced liver disease related to antituberculosis medications in East Asian population, but not the Indians or Caucasians[79]. There were no confirmed relationships between null genotype of GSTT1 gene and this kind of drug induced liver disease[79]. On the other hand, Wu et al[80] investigated that patients with tuberculosis A allele carriers of GSTP1 gene (A313G) have a higher risk of anti-tuberculosis drug-induced hepatotoxicity development.

LIVER CIRRHOSIS

With regards to the report of Burim et al[81] study of susceptibility to cirrhosis and pancreatitis in alcoholic, concerning the GST and cytochromes 450 genes polymorphism, revealed that GSTP1 Val allele carriers were at higher risk of both diseases. Ghobadloo et al[82] discovered the association of cryptogenic cirrhosis with Val/Val GSTP1 genotype which might be explained by low detoxification activity of protein that implicate this polymorphism as a risk factor for occurrence of the disease. Goncharova et al[83] showed that patients with liver cirrhosis AA genotype carriers have 2.5 times higher survival rate compared with the patients with the GG and AG genotypes of GSTP1 gene.

Khan et al[84] showed an increase in risk to alcoholic cirrhosis in patients with GSTM1 null genotype when compared with non-alcoholic or alcoholic controls. A much higher risk to alcoholic liver cirrhosis was observed in patients carrying combination of null genotypes of GSTM1 and GSTT1[84]. The authors of the mentioned study found interaction of GSTs with variant genotype of manganese superoxide dismutase, which detoxifies free radicals, or cytochrome P450 2E1 that generates free radicals, and resulted in several fold increase in risk to alcoholic liver cirrhosis. Thus, conclude the possible gene-gene interaction in modulating the risk of the alcoholic liver cirrhosis development[84]. However, in another study from Brazil, no differences were found in the prevalence of the GSTM1 and GSTT1 null genotypes between control non-alcoholics and alcoholics with liver cirrhosis, as well as alcoholics without disease and alcoholics with liver cirrhosis[81]. Several older studies also have got different conclusions regarding the impact of GSTM1 null genotype on the appearance of liver cirrhosis in patients with alcohol abuse. Specifically, Harada et al[85] in Japanese and Savolainen et al[86] in Finland found an increased risk of liver cirrhosis associated with the GSTM1 null genotype in chronic alcoholics. Whilst, Frenzer et al[87] in Caucasian population and Rodrigo et al[88] in Spanish adults have not reported any. Brind et al[89] have found higher prevalence of zero GSTT1 genotype in patients with alcoholic liver disease compared to patients who do not consume alcohol. Meanwhile GSTT1 null genotype was not found to vary importantly between liver cirrhosis related to hepatitis B infection and healthy individuals[90]. At the same time, 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[90]. Moreover, the link between GSTM1, but not GSTT1 null genotype and cryptogenic cirrhosis was found in Iranian population[82]. Komuro et al[91] in their investigations of primary biliary cirrhosis concluded that genotypic difference of GSTM1 and GSTT1 did not relate to susceptibility of this disease, nevertheless serum titer of anti-mitochondrial antibody of GSTM1 null and GSTT1 null patients were significantly higher than those of GSTM1 positive and/or GSTT1 positive patients. Baclig et al[92] also postulated that polymorphism in GSTM1 null genotype seems to be associated with an increased risk of chronic liver disease amongst Filipinos.

HEPATOCELLULAR CARCINOMA

The GST null genotype has been examined to have an association with various malignancies including cancers of the bladder[93], gastric[94], colon[95], and lung[96]. K. Wu et al[97] investigated that GSTP1 313 G/G polymorphism is a strong predisposing risk factor for bladder cancer. Meanwhile, data regarding the role of GST gene polymorphism on the hepatocellular carcinoma (HCC) is sporicidal. Qu et al[98] have found single nucleotide polymorphism (SNPs) GSTO2 rs7085725 and GSTP1 rs4147581 were significantly associated with the overall survival of HCC patients and suggested to use them alone or in combination as potential prognostic markers for HCC patients. Particularly, according to the author’s suggestion, SNP of GSTP1 (rs4147581) could have a predictive biomarker in HCC patients aged ≤ 55 years[98]. GSTM1 and GSTT1 polymorphisms appear to be associated with a modest increase in the risk of HCC in Egyptian patients[99]. GSTT1 null genotype was associated with more than 2-fold increased risk for HCC development in patients with hepatitis associated with hepatitis C virus (HCV) as compared to the control group. However, GSTM1 null genotype was found to have a protective effect when hepatitis patients were considered in Indian population[100]. Meanwhile, in older study it was found that the GSTT1-null genotype alone did not affect risk of HCC development in HBV, but the GSTM1-null genotype was associated with a decreased risk for early-onset HCC[101]. The meta-analysis by Li et al[102], involving results of 46 related studies with more than 15 thousands of patients showed that both GSTM1 null genotypes and GSTT1 null genotypes increased the risk of HCC, while GSTM1-GSTT1 dual-null genotypes increased the risk of HCC to a higher extend. Interestingly, during ethnicity consideration, this connection was significant only for Asians, and not for Caucasians and Africans. In older meta-analysis by Shen et al[103] GSTM1 and GSTT1 null genotype was found to be associated with higher risk of HCC with a similar ethnic pattern. GST-P1 rs1138272 (341C>T) polymorphism was found to have a protective effect on liver cancer development in a high-risk HCV/HBV-positive population in Caucasian ethnicity[104]. GST-P1 genetic polymorphisms (i.e., Ile105Val, rs1695) were not associated with HCC risk in Asian population, European and African[105,106]. Higher GSTP1 levels in tumor tissues indicated a better overall survival and disease-free survival for HCC patients[107]. The mentioned authors have found that GSTP1 could decrease p-Akt in liver cancer cell lines and may inhibit alfa-fetoprotein expression. GSTP1’s inhibition on cancer progression may be accomplished by arresting the cell cycle at the G1/S transition in HCC cells[108]. GSTA1 TT genotype was more frequent in HCC than in non-HCC patients, suggesting that individuals carrying this genotype could be associated with 2-fold higher risk of developing HCCs[109]. GSTM1 and GSTT1 null genotypes are associated with an increased HCC risk in Chinese population with higher risk typical for double null genotype. Furthermore, in another meta-analysis, it was investigated that null genotype of GSTT1 was associated with HCC susceptibility in Asians, and both GSTT1 and GSTM1 genes deletion were associated with higher susceptibility. GSTP1 Ile105 Val gene polymorphism was not correlated with this disease, however, polymorphisms in GSTM1 and GSTT1 genes are not related to the incidence of HCC in a high-risk Spanish population[110]. Marahatta et al[111] provided the support for the difference in genotypic distribution for GSTO1*A140D between hepatocellular carcinoma and cholangiocarcinoma.

CONCLUSION

Current review supports the position that genetic polymorphism of GST genes is involved in the pathogenesis of various liver diseases, specifically in non-alcoholic fatty liver disease, hepatitis and liver cirrhosis of different etiology and hepatocellular carcinoma. Certain GST gene allelic variants were proven to be associated with susceptibility to hepatological pathology and correlations with the natural course of the diseases were postulated. Still the data obtained in different studies sometimes is controversial and even conflicting. Thus, more investigations involving larger numbers of patients are needed.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Ukraine

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Jha RK, Pham TTT S-Editor: Zhang L L-Editor: A P-Editor: Wang LL

References
1.  Wilce MC, Parker MW. Structure and function of glutathione S-transferases. Biochim Biophys Acta. 1994;1205:1-18.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 458]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
2.  Fretland AJ, Omiecinski CJ. Epoxide hydrolases: biochemistry and molecular biology. Chem Biol Interact. 2000;129:41-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 203]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
3.  Hayes JD, Flanagan JU, Jowsey IR. Glutathione transferases. Annu Rev Pharmacol Toxicol. 2005;45:51-88.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2571]  [Cited by in F6Publishing: 2549]  [Article Influence: 134.2]  [Reference Citation Analysis (0)]
4.  Decker M, Arand M, Cronin A. Mammalian epoxide hydrolases in xenobiotic metabolism and signalling. Arch Toxicol. 2009;83:297-318.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 157]  [Cited by in F6Publishing: 141]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
5.  Chen CH  Phase II Enzymes. In: Chen CH. Activation and Detoxification Enzymes. New York: Springer, 2012: 37-48.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Çelįk SK, Aras N, Yildirim Ö, Turan F, Görür A, Yildirim H, Tamer L. Glutathione S-transferase GSTM 1, null genotype may be associated with susceptibility to age-related cataract. Adv Clin Exp Med. 2015;24:113-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
7.  Townsend DM, Tew KD. The role of glutathione-S-transferase in anti-cancer drug resistance. Oncogene. 2003;22:7369-7375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 864]  [Cited by in F6Publishing: 872]  [Article Influence: 41.5]  [Reference Citation Analysis (0)]
8.  Tew KD, Townsend DM. Glutathione-s-transferases as determinants of cell survival and death. Antioxid Redox Signal. 2012;17:1728-1737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 135]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
9.  La Roche SD, Leisinger T. Sequence analysis and expression of the bacterial dichloromethane dehalogenase structural gene, a member of the glutathione S-transferase supergene family. J Bacteriol. 1990;172:164-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 141]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
10.  Hayes JD, Pulford DJ. The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance. Crit Rev Biochem Mol Biol. 1995;30:445-600.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2409]  [Cited by in F6Publishing: 2364]  [Article Influence: 81.5]  [Reference Citation Analysis (0)]
11.  Sheehan D, Meade G, Foley VM, Dowd CA. Structure, function and evolution of glutathione transferases: implications for classification of non-mammalian members of an ancient enzyme superfamily. Biochem J. 2001;360:1-16.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 744]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
12.  Kovaleva J, Degener JE, van der Mei HC. Methylobacterium and its role in health care-associated infection. J Clin Microbiol. 2014;52:1317-1321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
13.  Gene group: Soluble glutathione S-transferases (GST);  [cited 20 March 2021].   Available from: https://www.genenames.org/data/genegroup/#!/group/567.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Nebert DW, Vasiliou V. Analysis of the glutathione S-transferase (GST) gene family. Hum Genomics. 2004;1:460-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 227]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
15.  Islam S, Sajib SD, Jui ZS, Arabia S, Islam T, Ghosh A. Genome-wide identification of glutathione S-transferase gene family in pepper, its classification, and expression profiling under different anatomical and environmental conditions. Sci Rep. 2019;9:9101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
16.  Marco A, Cuesta A, Pedrola L, Palau F, Marín I. Evolutionary and structural analyses of GDAP1, involved in Charcot-Marie-Tooth disease, characterize a novel class of glutathione transferase-related genes. Mol Biol Evol. 2004;21:176-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 69]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
17.  Saisawang C, Wongsantichon J, Ketterman AJ. A preliminary characterization of the cytosolic glutathione transferase proteome from Drosophila melanogaster. Biochem J. 2012;442:181-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
18.  Wongtrakul J, Janphen K, Saisawang C, Ketterman AJ. Interaction of Omega, Sigma, and Theta glutathione transferases with p38b mitogen-activated protein kinase from the fruit fly, Drosophila melanogaster. J Insect Sci. 2014;14:60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
19.  Schröder P, Scheer CE, Diekmann F, Stampfl A. How plants cope with foreign compounds. Translocation of xenobiotic glutathione conjugates in roots of barley (Hordeum vulgare). Environ Sci Pollut Res Int. 2007;14:114-122.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 18]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
20.  Dixon DP, Edwards R. Selective binding of glutathione conjugates of fatty acid derivatives by plant glutathione transferases. J Biol Chem. 2009;284:21249-21256.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 65]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
21.  Dixon DP, Davis BG, Edwards R. Functional divergence in the glutathione transferase superfamily in plants. Identification of two classes with putative functions in redox homeostasis in Arabidopsis thaliana. J Biol Chem. 2002;277:30859-30869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 326]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
22.  Dixon DP, Skipsey M, Edwards R. Roles for glutathione transferases in plant secondary metabolism. Phytochemistry. 2010;71:338-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 257]  [Article Influence: 18.4]  [Reference Citation Analysis (0)]
23.  Wagner U, Edwards R, Dixon DP, Mauch F. Probing the diversity of the Arabidopsis glutathione S-transferase gene family. Plant Mol Biol. 2002;49:515-532.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 331]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
24.  Landi S. Mammalian class theta GST and differential susceptibility to carcinogens: a review. Mutat Res. 2000;463:247-283.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 343]  [Cited by in F6Publishing: 359]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
25.  Katoh T, Inatomi H, Nagaoka A, Sugita A. Cytochrome P4501A1 gene polymorphism and homozygous deletion of the glutathione S-transferase M1 gene in urothelial cancer patients. Carcinogenesis. 1995;16:655-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 45]  [Cited by in F6Publishing: 48]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
26.  Hamajima N, Takezaki T, Tajima K. Allele Frequencies of 25 Polymorphisms Pertaining to Cancer Risk for Japanese, Koreans and Chinese. Asian Pac J Cancer Prev. 2002;3:197-206.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Coles BF, Kadlubar FF. Detoxification of electrophilic compounds by glutathione S-transferase catalysis: determinants of individual response to chemical carcinogens and chemotherapeutic drugs? Biofactors. 2003;17:115-130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 154]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
28.  Townsend D, Tew K. Cancer drugs, genetic variation and the glutathione-S-transferase gene family. Am J Pharmacogenomics. 2003;3:157-172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 105]  [Cited by in F6Publishing: 101]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
29.  Katoh T, Yamano Y, Tsuji M, Watanabe M. Genetic polymorphisms of human cytosol glutathione S-transferases and prostate cancer. Pharmacogenomics. 2008;9:93-104.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
30.  Kautiainen RJ, Dwivedi B, MacDonald TJ, King TZ. GSTP1 polymorphisms sex-specific association with verbal intelligence in survivors of pediatric medulloblastoma tumors. Child Neuropsychol. 2020;26:739-753.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
31.  Liang Q, He JS, Fulco AJ. The role of Barbie box sequences as cis-acting elements involved in the barbiturate-mediated induction of cytochromes P450BM-1 and P450BM-3 in Bacillus megaterium. J Biol Chem. 1995;270:4438-4450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 68]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
32.  Gullner G, Komives T, Király L, Schröder P. Glutathione S-Transferase Enzymes in Plant-Pathogen Interactions. Front Plant Sci. 2018;9:1836.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 193]  [Article Influence: 32.2]  [Reference Citation Analysis (0)]
33.  Wu JH, Batist G. Glutathione and glutathione analogues; therapeutic potentials. Biochim Biophys Acta. 2013;1830:3350-3353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
34.  Harshbarger W, Gondi S, Ficarro SB, Hunter J, Udayakumar D, Gurbani D, Singer WD, Liu Y, Li L, Marto JA, Westover KD. Structural and Biochemical Analyses Reveal the Mechanism of Glutathione S-Transferase Pi 1 Inhibition by the Anti-cancer Compound Piperlongumine. J Biol Chem. 2017;292:112-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 56]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
35.  Allocati N, Masulli M, Di Ilio C, Federici L. Glutathione transferases: substrates, inihibitors and pro-drugs in cancer and neurodegenerative diseases. Oncogenesis. 2018;7:8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 298]  [Article Influence: 49.7]  [Reference Citation Analysis (0)]
36.  Sappl PG, Carroll AJ, Clifton R, Lister R, Whelan J, Harvey Millar A, Singh KB. The Arabidopsis glutathione transferase gene family displays complex stress regulation and co-silencing multiple genes results in altered metabolic sensitivity to oxidative stress. Plant J. 2009;58:53-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 199]  [Cited by in F6Publishing: 174]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
37.  Liao W, Ji L, Wang J, Chen Z, Ye M, Ma H, An X. Identification of glutathione S-transferase genes responding to pathogen infestation in Populus tomentosa. Funct Integr Genomics. 2014;14:517-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
38.  Wahibah NN, Tsutsui T, Tamaoki D, Sato K, Nishiuchi T. Expression of barley Glutathione S-Transferase13 gene reduces accumulation of reactive oxygen species by trichothecenes and paraquat in Arabidopsis plants. Plant Biotechnol (Tokyo). 2018;35:71-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 9]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
39.  Hernandez JA, Barba-Espin G, Diaz-Vivancos P.   Glutathione-Mediated Biotic Stress Tolerance in Plants. In: Hossain MA, Mostofa MA, Vuvancos PD, Burritt DJ, Fujita M, Tran LSP. Glutathione in Plant Growth, Development, and Stress Tolerance, Springer International Publishing, 2017: 309-329.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Ouaissi A, Ouaissi M, Sereno D. Glutathione S-transferases and related proteins from pathogenic human parasites behave as immunomodulatory factors. Immunol Lett. 2002;81:159-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
41.  Bae YA, Kim JG, Kong Y. Phylogenetic characterization of Clonorchis sinensis proteins homologous to the sigma-class glutathione transferase and their differential expression profiles. Mol Biochem Parasitol. 2016;206:46-55.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 11]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
42.  Higgins LG, Hayes JD. Mechanisms of induction of cytosolic and microsomal glutathione transferase (GST) genes by xenobiotics and pro-inflammatory agents. Drug Metab Rev. 2011;43:92-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 138]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
43.  Dayama G, Priya S, Niccum DE, Khoruts A, Blekhman R. Interactions between the gut microbiome and host gene regulation in cystic fibrosis. Genome Med. 2020;12:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
44.  Sydorchuk LP, Sokolenko AA, Sydorchuk AR, Kryklyvets LG, Biryuk IG, Fliundra IG, Sokolenko MA. Insulin resistance in patients with arterial hypertension and abdominal obesity depending on ACE and PPAR-γ2 genes polymorphism: A new opinion concerning an old problem. New Armenian Med J. 2015;9:43-51.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Boušová I, Skálová L. Inhibition and induction of glutathione S-transferases by flavonoids: possible pharmacological and toxicological consequences. Drug Metab Rev. 2012;44:267-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 44]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
46.  Dixon DP, Sellars JD, Edwards R. The Arabidopsis phi class glutathione transferase AtGSTF2: binding and regulation by biologically active heterocyclic ligands. Biochem J. 2011;438:63-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
47.  Csiszár J, Horváth E, Váry Z, Gallé Á, Bela K, Brunner S, Tari I. Glutathione transferase supergene family in tomato: Salt stress-regulated expression of representative genes from distinct GST classes in plants primed with salicylic acid. Plant Physiol Biochem. 2014;78:15-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 82]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
48.  Tian M, von Dahl CC, Liu PP, Friso G, van Wijk KJ, Klessig DF. The combined use of photoaffinity labeling and surface plasmon resonance-based technology identifies multiple salicylic acid-binding proteins. Plant J. 2012;72:1027-1038.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
49.  Sydorchuk L, Yarynych Y, Knut R, Sydorchuk A, Matyukha L, Sydorchuk O, Fedoniuk L, Khodorovskyi V, Petrynych V, Babinec L, Reva T, Sydorchuk R. Hepatocytes' function and adipokines in patients with non-alcoholic fatty liver disease depending on the ACE (rs4646994) and PPAR-γ2 (rs1801282) genes' polymorphisms. Rev Med Chir Soc Med Nat Iaşi. 2018;122:358-364.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Prysyazhnyuk VP, Rossokha ZI, Gorovenko NG. Variation in particular biochemical indicators, cytokine and adipokine profiles of the blood, and the structural and functional parameters of the liver in patients with nonalcoholic fatty liver disease and different genotypes by the polymorphic locus A313G of the GSTP1 gene. Cytol Genet. 2017;6:50-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
51.  Hashemi M, Eskandari-Nasab E, Fazaeli A, Bahari A, Hashemzehi NA, Shafieipour S, Taheri M, Moazeni-Roodi A, Zakeri Z, Bakhshipour A, Ghavami S. Association of genetic polymorphisms of glutathione-S-transferase genes (GSTT1, GSTM1, and GSTP1) and susceptibility to nonalcoholic fatty liver disease in Zahedan, Southeast Iran. DNA Cell Biol. 2012;31:672-677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
52.  Prysyazhnyuk VP, Sydorchuk LP. Association of A313G polymorphism of GSTP1 gene with biochemical blood parameters in patients with nonalcoholic fatty liver disease. Archives of the Balkan Medical Union. 2016;51:178-182.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Liedtke C, Trautwein C. The role of TNF and Fas dependent signaling in animal models of inflammatory liver injury and liver cancer. Eur J Cell Biol. 2012;91:582-589.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 60]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
54.  Brenner C, Galluzzi L, Kepp O, Kroemer G. Decoding cell death signals in liver inflammation. J Hepatol. 2013;59:583-594.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 583]  [Cited by in F6Publishing: 650]  [Article Influence: 59.1]  [Reference Citation Analysis (0)]
55.  Younossi ZM, Jarrar M, Nugent C, Randhawa M, Afendy M, Stepanova M, Rafiq N, Goodman Z, Chandhoke V, Baranova A. A novel diagnostic biomarker panel for obesity-related nonalcoholic steatohepatitis (NASH). Obes Surg. 2008;18:1430-1437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 189]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
56.  Li G, Hu H, Shi W, Li Y, Liu L, Chen Y, Hu X, Wang J, Gao J, Yin D. Elevated hematocrit in nonalcoholic fatty liver disease: a potential cause for the increased risk of cardiovascular disease? Clin Hemorheol Microcirc. 2012;51:59-68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
57.  Paniagua JA. Nutrition, insulin resistance and dysfunctional adipose tissue determine the different components of metabolic syndrome. World J Diabetes. 2016;7:483-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 94]  [Cited by in F6Publishing: 93]  [Article Influence: 11.6]  [Reference Citation Analysis (2)]
58.  Altirriba J, Poher AL, Rohner-Jeanrenaud F. Chronic Oxytocin Administration as a Treatment Against Impaired Leptin Signaling or Leptin Resistance in Obesity. Front Endocrinol (Lausanne). 2015;6:119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 38]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
59.  Kaser S, Moschen A, Cayon A, Kaser A, Crespo J, Pons-Romero F, Ebenbichler CF, Patsch JR, Tilg H. Adiponectin and its receptors in non-alcoholic steatohepatitis. Gut. 2005;54:117-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 323]  [Cited by in F6Publishing: 300]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
60.  Perumpail RB, Liu A, Wong RJ, Ahmed A, Harrison SA. Pathogenesis of hepatocarcinogenesis in non-cirrhotic nonalcoholic fatty liver disease: Potential mechanistic pathways. World J Hepatol. 2015;7:2384-2388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
61.  Hori M, Oniki K, Nakagawa T, Takata K, Mihara S, Marubayashi T, Nakagawa K. Association between combinations of glutathione-S-transferase M1, T1 and P1 genotypes and non-alcoholic fatty liver disease. Liver Int. 2009;29:164-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
62.  Prysyazhnyuk V, Voloshyn O, Prysiazhniuk I, Ilashchuk T, Sydorchuk L, Prysyazhnyuk P. Glutathione S-transferase T1 and M1 null genotype distribution among non-alcoholic fatty liver disease patients and its association with cytokine and adipokine profiles. Clin Exp Hepatol. 2020;6:142-149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
63.  Rafiee L, Shokouh P, Roohafza H, Mansourian M, Javanmard SH. Association of glutathione S-transferases M1 and T1 gene polymorphisms with the risk of metabolic syndrome in an Iranian population. Adv Biomed Res. 2016;5:63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
64.  Maciel SS, Pereira Ada C, Silva GJ, Rodrigues MV, Mill JG, Krieger JE. Association between glutathione S-transferase polymorphisms and triglycerides and HDL-cholesterol. Atherosclerosis. 2009;206:204-208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 34]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
65.  Chakraborty A, Choudhury A, Saha A. Development of Non-alcoholic Fatty Liver Disease (NAFLD) in Young Obese Tribal Subjects of Tripura: Link between Low 25 (OH) Vitamin-D Levels and Immune Modulators. J Assoc Physicians India. 2019;67:52-56.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Jamali R, Arj A, Razavizade M, Aarabi MH. Prediction of Nonalcoholic Fatty Liver Disease Via a Novel Panel of Serum Adipokines. Medicine (Baltimore). 2016;95:e2630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 51]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
67.  Polyzos SA, Kountouras J, Zavos Ch. The multi-hit process and the antagonistic roles of tumor necrosis factor-alpha and adiponectin in non alcoholic fatty liver disease. Hippokratia. 2009;13:127; author reply 128.  [PubMed]  [DOI]  [Cited in This Article: ]
68.  Prysyazhnyuk VP, Sydorchuk LP. The relationship between GSTT1 and GSTM1 deletion gene polymorphisms with biochemical blood parameters, indicators of pro- and antioxidant systems in patients with nonalcoholic fatty liver disease. Gastro. 2015;5:40-47.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Prysyazhnyuk VP, Voloshyn OI, Sydorchuk LP. Association of A313G polymorphism of GSTP1 gene with biochemical, pro- and antioxidant blood indicators in chronic hepatitis patients. J Pharm Innov. 2017;6:40-43a.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Kandemir O, Tamer L, Tasdelen B. Effects of GSTT1, GSTM1 and GSTP1 gene polymorphism on the course of hepatitis B virus infection. Hepatogastroenterology. 2008;55:1729-1733.  [PubMed]  [DOI]  [Cited in This Article: ]
71.  Qi L, Zou ZQ, Wang LY, Gao S, Fan YC, Long B, Guo YM, Xu AL, Han J, Li T, Wang K. Methylation of the glutathione-S-transferase M3 gene promoter is associated with oxidative stress in acute-on-chronic hepatitis B liver failure. Tohoku J Exp Med. 2012;228:43-51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
72.  Shaban NZ, Salem HH, Elsadany MA, Ali BA, Hassona EM, Mogahed FA. Distribution of Glutathione S-Transferase Omega Gene Polymorphism with Different Stages of HBV Infection Including Hepatocellular Carcinoma in the Egyptian Population. Asian Pac J Cancer Prev. 2016;17:2145-2150.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
73.  Roy B, Chowdhury A, Kundu S, Santra A, Dey B, Chakraborty M, Majumder PP. Increased risk of antituberculosis drug-induced hepatotoxicity in individuals with glutathione S-transferase M1 'null' mutation. J Gastroenterol Hepatol. 2001;16:1033-1037.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 135]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
74.  Leiro V, Fernández-Villar A, Valverde D, Constenla L, Vázquez R, Piñeiro L, González-Quintela A. Influence of glutathione S-transferase M1 and T1 homozygous null mutations on the risk of antituberculosis drug-induced hepatotoxicity in a Caucasian population. Liver Int. 2008;28:835-839.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 77]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
75.  Monteiro TP, El-Jaick KB, Jeovanio-Silva AL, Brasil PE, Costa MJ, Rolla VC, de Castro L. The roles of GSTM1 and GSTT1 null genotypes and other predictors in anti-tuberculosis drug-induced liver injury. J Clin Pharm Ther. 2012;37:712-718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
76.  Chatterjee S, Lyle N, Mandal A, Kundu S. GSTT1 and GSTM1 gene deletions are not associated with hepatotoxicity caused by antitubercular drugs. J Clin Pharm Ther. 2010;35:465-470.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
77.  Ikeda T. Drug-induced idiosyncratic hepatotoxicity: prevention strategy developed after the troglitazone case. Drug Metab Pharmacokinet. 2011;26:60-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
78.  Perwitasari DA, Atthobari J, Wilffert B. Pharmacogenetics of isoniazid-induced hepatotoxicity. Drug Metab Rev. 2015;47:222-228.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
79.  Huang YS. Recent progress in genetic variation and risk of antituberculosis drug-induced liver injury. J Chin Med Assoc. 2014;77:169-173.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 39]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
80.  Wu S, Wang YJ, Tang X, Wang Y, Wu J, Ji G, Zhang M, Chen G, Liu Q, Sandford AJ, He JQ. Genetic Polymorphisms of Glutathione S-Transferase P1 (GSTP1) and the Incidence of Anti-Tuberculosis Drug-Induced Hepatotoxicity. PLoS One. 2016;11:e0157478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
81.  Burim RV, Canalle R, Martinelli Ade L, Takahashi CS. Polymorphisms in glutathione S-transferases GSTM1, GSTT1 and GSTP1 and cytochromes P450 CYP2E1 and CYP1A1 and susceptibility to cirrhosis or pancreatitis in alcoholics. Mutagenesis. 2004;19:291-298.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 71]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
82.  Ghobadloo SM, Yaghmaei B, Bakayev V, Goudarzi H, Noorinayer B, Rad FH, Samiy S, Aghabozorghi S, Zali MR. GSTP1, GSTM1, and GSTT1 genetic polymorphisms in patients with cryptogenic liver cirrhosis. J Gastrointest Surg. 2004;8:423-427.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
83.  Goncharova IA, Rachkovskiĭ MI, Beloborodova EV, Gamal' Abd El'-Aziz Nasar Kh, Puzyrev VP. [Liver cirrhosis patogenetics: polymorphism of glutation S-transferase genes]. Mol Biol (Mosk). 2010;44:431-438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
84.  Khan AJ, Choudhuri G, Husain Q, Parmar D. Polymorphism in glutathione-S-transferases: a risk factor in alcoholic liver cirrhosis. Drug Alcohol Depend. 2009;101:183-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
85.  Harada S, Takase S, Horiike N, Ishii K, Ishii H, Takada A. [Genetic and epidemiologic studies on alcoholic liver diseases]. Arukoru Kenkyuto Yakubutsu Ison. 1993;28:400-413.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Savolainen VT, Pjarinen J, Perola M, Penttilä A, Karhunen PJ. Glutathione-S-transferase GST M1 "null" genotype and the risk of alcoholic liver disease. Alcohol Clin Exp Res. 1996;20:1340-1345.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 33]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
87.  Frenzer A, Butler WJ, Norton ID, Wilson JS, Apte MV, Pirola RC, Ryan P, Roberts-Thomson IC. Polymorphism in alcohol-metabolizing enzymes, glutathione S-transferases and apolipoprotein E and susceptibility to alcohol-induced cirrhosis and chronic pancreatitis. J Gastroenterol Hepatol. 2002;17:177-182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 65]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
88.  Rodrigo L, Alvarez V, Rodriguez M, Pérez R, Alvarez R, Coto E. N-acetyltransferase-2, glutathione S-transferase M1, alcohol dehydrogenase, and cytochrome P450IIE1 genotypes in alcoholic liver cirrhosis: a case-control study. Scand J Gastroenterol. 1999;34:303-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
89.  Brind AM, Hurlstone A, Edrisinghe D, Gilmore I, Fisher N, Pirmohamed M, Fryer AA. The role of polymorphisms of glutathione S-transferases GSTM1, M3, P1, T1 and A1 in susceptibility to alcoholic liver disease. Alcohol Alcoholism. 2004;39:478-483.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
90.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 5]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
91.  Komuro O, Takahashi H, Sato K, Tamaki S, Zeniya M, Toda G. [Significance of serum oxidative stress related markers and genotype of GST gene in the pathogeneses of primary biliary cirrhosis]. Nihon Rinsho Meneki Gakkai Kaishi. 2004;27:322-329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
92.  Baclig MO, Alvarez MR, Lozada XM, Mapua CA, Lozano-Kühne JP, Dimamay MP, Natividad FF, Gopez-Cervantes J, Matias RR. Association of glutathione S-transferase T1 and M1 genotypes with chronic liver diseases among Filipinos. Int J Mol Epidemiol Genet. 2012;3:153-159.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Bell DA, Taylor JA, Paulson DF, Robertson CN, Mohler JL, Lucier GW. Genetic risk and carcinogen exposure: a common inherited defect of the carcinogen-metabolism gene glutathione S-transferase M1 (GSTM1) that increases susceptibility to bladder cancer. J Natl Cancer Inst. 1993;85:1159-1164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 444]  [Cited by in F6Publishing: 467]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
94.  Wang ZY, Zhou J, Luo L, Huang YL, Dong PD. Predictive role of glutathione-S-transferase gene polymorphisms in the survival of gastric cancer cases. Asian Pac J Cancer Prev. 2012;13:1515-1518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
95.  Welfare M, Monesola Adeokun A, Bassendine MF, Daly AK. Polymorphisms in GSTP1, GSTM1, and GSTT1 and susceptibility to colorectal cancer. Cancer Epidemiol Biomarkers Prev. 1999;8:289-292.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Dialyna IA, Miyakis S, Georgatou N, Spandidos DA. Genetic polymorphisms of CYP1A1, GSTM1 and GSTT1 genes and lung cancer risk. Oncol Rep. 2003;10:1829-1835.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Wu K, Wang X, Xie Z, Liu Z, Lu Y. Glutathione S-transferase P1 gene polymorphism and bladder cancer susceptibility: an updated analysis. Mol Biol Rep. 2013;40:687-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 15]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
98.  Qu K, Liu SS, Wang ZX, Huang ZC, Liu SN, Chang HL, Xu XS, Lin T, Dong YF, Liu C. Polymorphisms of glutathione S-transferase genes and survival of resected hepatocellular carcinoma patients. World J Gastroenterol. 2015;21:4310-4322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
99.  Abd El-Moneim E, Younis FA, Allam N, Gameel K, Osman M. Gene deletion of glutathione S-transferase M1 and T1 and risk factors of hepatocellular carcinoma in Egyptian patients. Egypt J Immunol. 2008;15:125-134.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Kiran M, Chawla YK, Kaur J. Glutathione-S-transferase and microsomal epoxide hydrolase polymorphism and viral-related hepatocellular carcinoma risk in India. DNA Cell Biol. 2008;27:687-694.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
101.  Yu MW, Yang SY, Pan IJ, Lin CL, Liu CJ, Liaw YF, Lin SM, Chen PJ, Lee SD, Chen CJ. Polymorphisms in XRCC1 and glutathione S-transferase genes and hepatitis B-related hepatocellular carcinoma. J Natl Cancer Inst. 2003;95:1485-1488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 82]  [Cited by in F6Publishing: 88]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
102.  Li S, Xue F, Zheng Y, Yang P, Lin S, Deng Y, Xu P, Zhou L, Hao Q, Zhai Z, Wu Y, Dai Z, Chen S. GSTM1 and GSTT1 null genotype increase the risk of hepatocellular carcinoma: evidence based on 46 studies. Cancer Cell Int. 2019;19:76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
103.  Shen YH, Chen S, Peng YF, Shi YH, Huang XW, Yang GH, Ding ZB, Yi Y, Zhou J, Qiu SJ, Fan J, Ren N. Quantitative assessment of the effect of glutathione S-transferase genes GSTM1 and GSTT1 on hepatocellular carcinoma risk. Tumour Biol. 2014;35:4007-4015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
104.  De Mattia E, Cecchin E, Polesel J, Bignucolo A, Roncato R, Lupo F, Crovatto M, Buonadonna A, Tiribelli C, Toffoli G. Genetic biomarkers for hepatocellular cancer risk in a caucasian population. World J Gastroenterol. 2017;23:6674-6684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 21]  [Cited by in F6Publishing: 18]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
105.  Chen J, Ma L, Peng NF, Wang SJ, Li LQ. A meta-analysis of the relationship between glutathione S-transferases gene polymorphism and hepatocellular carcinoma in Asian population. Mol Biol Rep. 2012;39:10383-10393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
106.  White DL, Li D, Nurgalieva Z, El-Serag HB. Genetic variants of glutathione S-transferase as possible risk factors for hepatocellular carcinoma: a HuGE systematic review and meta-analysis. Am J Epidemiol. 2008;167:377-389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 92]  [Cited by in F6Publishing: 93]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
107.  Liu X, Tan N, Liao H, Pan G, Xu Q, Zhu R, Zou L, He S, Zhu H. High GSTP1 inhibits cell proliferation by reducing Akt phosphorylation and is associated with a better prognosis in hepatocellular carcinoma. Oncotarget. 2018;9:8957-8971.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
108.  Liu K, Zhang L, Lin X, Chen L, Shi H, Magaye R, Zou B, Zhao J. Association of GST genetic polymorphisms with the susceptibility to hepatocellular carcinoma (HCC) in Chinese population evaluated by an updated systematic meta-analysis. PLoS One. 2013;8:e57043.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
109.  Akhdar H, El Shamieh S, Musso O, Désert R, Joumaa W, Guyader D, Aninat C, Corlu A, Morel F. The rs3957357C>T SNP in GSTA1 Is Associated with a Higher Risk of Occurrence of Hepatocellular Carcinoma in European Individuals. PLoS One. 2016;11:e0167543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
110.  Ladero JM, Martínez C, García-Martín E, Ropero P, Briceño O, Villegas A, Díaz-Rubio M, Agúndez JA. Glutathione S-transferase M1 and T1 genetic polymorphisms are not related to the risk of hepatocellular carcinoma: a study in the Spanish population. Eur J Cancer. 2006;42:73-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
111.  Marahatta SB, Punyarit P, Bhudisawasdi V, Paupairoj A, Wongkham S, Petmitr S. Polymorphism of glutathione S-transferase omega gene and risk of cancer. Cancer Lett. 2006;236:276-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 80]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]