Figure 4 - uploaded by Libia Vega
Content may be subject to copyright.
Schematic representation of cis -acting elements involved in the transcriptional regulation of the GST Ya subunit gene. cis -acting regulatory elements identified in the 5 ́-flanking region of the GST Ya subunit gene promoter, HNF1 (nucleotides -860 to - 850) and HFN4 (nucleotides -775 to -755); GRE (nucleotides -1609 to -1595) (Adapted from 3). 

Schematic representation of cis -acting elements involved in the transcriptional regulation of the GST Ya subunit gene. cis -acting regulatory elements identified in the 5 ́-flanking region of the GST Ya subunit gene promoter, HNF1 (nucleotides -860 to - 850) and HFN4 (nucleotides -775 to -755); GRE (nucleotides -1609 to -1595) (Adapted from 3). 

Source publication
Chapter
Full-text available
Glutathione S-transferases (GSTs) are a family of the phase II metabolic enzymes that catalyze the nucleophilic attack of the sulfur atom of glutathione (gamma-glutamyl-cysteinylglycine) to an electrophilic group on metabolic products or xenobiotic compounds. There are 8 types of GSTs with wide distribution within the organisms and redundant as wel...

Context in source publication

Context 1
... S-conjugate formation, translocation of the glutathione S-conjugates or the corresponding cysteine S-conjugates to the kidney and renal activation by cysteine conjugate β -lyase (21). In the kidney, cysteine S-conjugates are taken up by amino acid transport systems and undergo β -lyase-catalyzed metabolism. β -lyases a pyridoxal phosphate-dependent enzyme catalyzes β -elimination reactions of cysteine S-conjugates to yield ammonia, pyruvate and -chlorovinyl- or fluoroalkylthiolates as products. In cancer chemotherapy, the ability of GSTs to produce reactive metabolites has been exploited to target tumors that over express particular transferases (24), such as the latent cytotoxic drug TER286 (now called TLK286) (25) and the prodrug PABA/NO (O2-[2,4-dinitro-5-(N-methyl-N-4-carboxyphenylamino)phenyl] 1-N,N-dimethylamino)diazen-1-ium-1,2-diolate) (26). The process of aerobic respiration can lead to the production of reactive oxygen species (super oxide anion O 2 − , hydrogen peroxide H 2 O 2 , and the hydroxyl radical • HO) (27). Free radicals primarily arise through oxidative phosphorylation, although 5-lipoxygenase-, cyclooxygenase-, cytochrome P450-, and xanthene oxidase–catalyzed reactions are also a source. Such species are scavenged by the catalytic activities of superoxide dismutase, catalase, and GPx and nonenzymatically by α -tocopherol, ascorbic acid, GSH, and bilirubin. Despite these antioxidant defenses, reactive oxygen species inflict damage on membrane lipid, DNA, protein, and carbohydrate. The GSTs both soluble and MAPEG supergene families have the capacity to reduce these and another compounds (Table 3), because they also display GPx activity, and thus protect from oxidative damage (13), for example, they can detoxify downstream products of oxidative damage such as the reactive aldehydes, 4-hydroxynonenal and acrolein (9). The GSTZ has been identified as the maleylacetoacetate isomerase that catalyzes the penultimate step in the catabolism of phenylalanine and tyrosine (7), where phenylalanine is degraded to acetoacetate and fumaric acid. The five intermediates are tyrosine, 4-hydroxyphenylpyruvate, homogentisate, aleyacetoacetate and fumarylacetoacetate. Mammal GSTs are involved in the intracellular transport of a variety of endogenous metabolites, drugs, and hormones. Particularly, GSTs are glucocorticoid-binding proteins and, thereby, may influence transport, metabolism, and action of steroids. The GSTs also participate in the intracellular binding and transport of a broad range of other lipophilic compounds including bilirubin, glucocorticoids and thyroid hormones. It has been demonstrated that testosterone and progesterone have the ability to bind GSTs with moderate affinity. Thus, GSTs could play a key role in the physiological action of these hormones (29). The metabolic pathways of steroid hormone biosynthesis leading to compounds such as testosterone and progesterone start with cholesterol and proceed in multiple steps involving oxidation and isomerization reactions that are mediated by GSTA3-3 (Figure 3) (28). The GSTA3-3 is expressed in steroidogenic tissues like testis, ovary, adrenal gland, and placenta. The GSTA1-1 is a major hepatic enzyme (2-3% of the total cytosolic protein) and is also found in significant amounts in kidney and testis. In human gonads, GSTA have been immunohistochemically identified in cells involved in hormone production, i.e. testicular interstitial Leydig cells and ovarian cells of the Graffian follicle and corpus luteum as well as cells in the reticular layer of the adrenal cortex. This selective tissue expression provides support for a role of GSTs in steroid hormone biosynthesis (28). On the other hand, GSTs contribute to the biosynthesis of pharmacologically important metabolites of arachidonic acid (7). Membrane-associated GSTs and related proteins have been linked to the metabolism of eicosanoids such as leukotrienes and prostaglandins, however, several of the soluble enzymes such as GSTA2-2, GSTM2-2, and GSTM3-3 also display activity of physiological relevance with prostaglandins (28). The regulation of cytosolic GSTs is subjected to a complex set of endogenous parameters. These include development, sex and tissue specific factors, as well as a large number of xenobiotics, inducing agents such as PAHs, phenolic antioxidants, Michael acceptors, reactive oxygen species, isothiocyanates, trivalents arsenicals, barbiturates and synthetic glucocorticoids (6). Eaton and Bammler (30), describe that the majority of the GSTs are regulated by glucocorticoid-response elements (GRE), xenobiotic response elements (XRE), and antioxidant-response elements (ARE) (Figure 3). Thus, induction of GSTs by dexamethasone, 3-methyl-chloranthene, dioxins, hydroxyanisole (BHA), butylated hidroxytoluene (BHT), and oltipraz has been described (31). Okuda et al. (32) characterized the rat P (Yp) gene; it contains 7 exons and 6 introns and spans approximately 3 kilobase pairs (Figure 4). Cowell et al. (33) and Morrow et al. (34) had characterized the human GSTM gene. Analysis of the human GSTT promoter revealed four putative transcription regulatory motifs. These sequences included a TATA box, 29 base pairs upstream from the start of transcription, two Spl recognition sequences (GGGCGG) at nucleotides -46 to -41 and -56 to -51 and an AP-1 recognition sequence (TGACTCA) at nucleotide position -69 to -63. Comparison of the human GSTM gene with the homologous rat gene disclosed extensive conservation of genomic organization between the two species (3). Functional AP-1 and SP1 response elements have also been identified in the 5 ́regulatory region of human GSTP1 gene. Post-transcriptional mechanisms such as mRNA stability are also involved in regulating human GSTP1-1 protein levels (35). Also, the expression of human GSTP1 may be influenced by the methylation status of a CpG island in the regulatory region of the gene (36). Also, the cytosolic GSTs and GSH synthesis is up-regulated by electrophiles through the Nrf2 transcription factor via the cis-acting ARE (indeed, also called electrophile response element) (13). Thimmulappa et al. (37), found several Nrf2- dependent genes coding for isozymes of GSTs, including GSTM and GSTA. The regulation of GSTM1 can be activated by electrophiles that react with cysteine 49, it was initially suggested to be particularly reactive (13). The notion that Nrf2 mediates basal expression of GSTs by endogenous thiol-active endobiotics is supported by the fact that in null mice of this transcription factor the normal homeostatic levels of many A, M, and P transferases are reduced (37). Variation in GSTs alleles is very common in the population and will presumably make a significant contribution to inter-individual differences in drug metabolism. Although cytosolic GST enzymes have a key position in biological detoxification processes, two of the most relevant human isoenzymes, GSTT1-1 and GSTM1-1, are genetically deleted (non-functional alleles GSTT1*0 and GSTM1*0) (38) in a high percentage of the human population with major ethnic differences. GSTT1-1 is highly conserved during evolution. There are three alleles in GSTM1 locus: GSTM1*A, GSTM1*B and GSTM1*0, this last allele corresponds to a gene deletion and homozygotes express no protein (Table 4) (39). GSTM1*A and GSTM1*B differ by one base in exon 7, the catalytic properties of the enzymes encoded by these alleles are similar. The genetic polymorphisms of the GSTP1 were describe in 1997 by Ali- Osman et al. (40), the wild type genotype is known as GSTP1*A, there are three alleles of GSTP1 (Table 4) with not known effects. The first hints about the existence of a polymorphic glutathione S- transferase Theta came from Peter et al. (41) demonstrating that erythrocytes from only 60% of individuals (“conjugators”) catalyze the conjugation of methyl chloride with glutathione while remaining 40% (“non-conjugators”) lack this activity. There are two genes in GST Theta family –GSTT1 and GSTT2- are located on chromosome 22 and are separated by about 50 kb. Like GSTM1, the GSTT1 locus has a deleted express no protein (42). More recently, Alexandrie et al. (43) found a novel functional polymorphism in the GSTT1 gene. Sequencing of GSTT1 cDNA revealed a single nucleotide substitution, 310A>C, that altered the amino acid residue 104 from threonine to proline (T104P). Modeling studies of GSTT1 have suggested that residue 104 is located in the middle of α -helix 4. Introduction of a α -helix-disrupting proline most likely distorts the conformation of the protein. Individuals that lacked GSTT1 activity and carried the variant allele, tentatively denoted GSTT1*B, had no detectable GSTT1 immunoreactive protein. Pemble et al. (42) showed that the GSTT1 gene was absent around 38% of the population. The presence or absence of the gene was coincident with the conjugator (GSTT1+) and nonconjugator (GSTT1-) phenotypes, respectively. Genotoxic effects such as induction of sister chromatid exchanges (SCE) after exposure of human blood to methyl bromide and other agents in vitro were found to be more pronounced in nonconjugators. Even the background levels of SCE were higher in the nonconjugator phenotype (44). In this aspect, we realized some experiments in human lymphocytes in vitro exposed to different concentrations of DCM; we found that high conjugators had more SCE that the low conjugators (Figure 5), showing that GSTT1 could be a susceptibility factor when humans are environmentally or occupationally exposed to dihalomethanes (45). The genetic frequency of these polymorphic proteins (GSTM1 and GSTT1) is variable between ethnic groups (Table 5). GSTM1 is inherited as autosomal dominant and between 40 and 60% of most populations express GSTM1. GSTT1-1 is also polymorphic in humans, with 20% of Caucasians and 80% of Asians lacking the enzyme. Recently we have detected that the frequency of the null allele for GSTT1 in ...

Similar publications

Article
Full-text available
Cellular decision making is based on regulatory circuits that associate signal thresholds to specific physiological actions. This transmission of information is subjected to molecular noise what can decrease its fidelity. Here, we show instead how such intrinsic noise enhances information transfer in the presence of multiple circuit copies. The res...
Article
Full-text available
Isozyme-specific post-translational regulation fine tunes signaling events. However, redundancy in sequence or activity renders links between isozyme-specific modifications and downstream functions uncertain. Methods to study this phenomenon are underdeveloped. Here we use a redox-targeting screen to reveal that Akt3 is a first-responding isozyme s...
Article
Full-text available
Ca2+ ions play a key role in a wide variety of environmental responses and developmental processes in plants, and several protein families with Ca2+-binding domains have evolved to meet these needs, including calmodulin (CaM) and calmodulin-like proteins (CMLs). These proteins have no catalytic activity, but rather act as sensor relays that regulat...
Article
Full-text available
Biased signaling or functional selectivity occurs when a 7TM-receptor preferentially activates one of several available pathways. It can be divided into three distinct forms: ligand bias, receptor bias, and tissue or cell bias, where it is mediated by different ligands (on the same receptor), different receptors (with the same ligand), or different...
Article
Full-text available
Unlabelled: Until today, there is no systemic treatment available for advanced cholangiocarcinoma (CCA). Recent studies have shown a frequent upregulation of the PI3K-AKT-mTOR and RAF-MEK-ERK pathways in this type of cancer. However, considering their high extend of redundancy and cross-talk, targeting only one pathway is likely to result in thera...

Citations

... 17 Three glutathione S-transferase (GST) genes (GSTP1, GSTT1, and GSTM1) code the GST proteins, which are essential for GC elimination by making its first step, conjugation, possible. 18,19 Nullallele variants of GSTM1 and GSTT1 caused by a deletion of the gene, result in the absence of activity of these enzymes. Additionally, it was reported that GSTP1 gene variants rs1695 (c.313A>G, p.Ile105Val) and rs1138272 (c.341C>T, p.Ala114Val) influence the activity and the structure of GSTP1 and alter the efficiency of GC conjugation, if the minor alleles are present. ...
Article
Full-text available
Background Response to glucocorticoid (GC) monotherapy in the initial phase of remission induction treatment in childhood acute lymphoblastic leukemia (ALL) represents important biomarker of prognosis and outcome. We aimed to study variants in several pharmacogenes ( NR3C1 , GSTs and ABCB1 ) that could contribute to improvement of GC response through personalization of GC therapy. Methods Retrospective study enrolling 122 ALL patients was carried out to analyze variants of NR3C1 (rs33389, rs33388 and rs6198), GSTT1 (null genotype), GSTM1 (null genotype), GSTP1 (rs1695 and rs1138272) and ABCB1 (rs1128503, rs2032582 and rs1045642) genes using PCR-based methodology. The marker of GC response was blast count per microliter of peripheral blood on treatment day 8. We carried out analysis in which cut-off value for GC response was 1000 (according to Berlin-Frankfurt-Munster [BFM] protocol), as well as 100 or 0 blasts per microliter. Results Carriers of rare NR3C1 rs6198 GG genotype were more likely to have blast count over 1000, than the non-carriers (p = 0.030). NR3C1 CAA (rs33389-rs33388-rs6198) haplotype was associated with blast number below 1000 (p = 0.030). GSTP1 GC haplotype carriers were more likely to have blast number below 1000 (p = 0.036), below 100 (p = 0.028) and to be blast negative (p = 0.054), while GSTP1 GT haplotype and rs1138272 T allele carriers were more likely to be blasts positive (p = 0.034 and p = 0.024, respectively). ABCB1 CGT (rs1128503-rs2032582-rs1045642) haplotype carriers were more likely to be blast positive (p = 0.018). Conclusions Our results have shown that NR3C1 rs6198 variant and GSTP1 rs1695-rs1138272 haplotype are the most promising pharmacogenomic markers of GC response in ALL patients.
... Variation in the GSTT1 allele is common in the population and presumably makes a significant contribution to inter-individual differences in drug metabolism or disease incidence. It has been reported that the variability of the most studied GST genes is associated with the risk of developing some types of cancer (Landi, 2000;Olvera-Bello and Vega, 2010). The GST gene is associated with the development of oesophageal carcinoma, and the combination with other GSTs or CYP450 s polymorphisms can increase the risk of developing other types of cancer or diseases. ...
Article
The carcinogenicity of dichloromethane (DCM) has been demonstrated by mutagenicity studies using bacteria and yeasts and using animal bioassays. Epidemiological studies indicate that exposure to DCM increases the incidences of liver and pancreas cancers. In the present study, we determine whether DCM generates DNA damage in human peripheral blood mononuclear cells (PBMCs) and whether that process depends on glutathione S-transferase theta (GSTT)-1 activity. GSTT1 is one of the enzymes that biotransforms DCM. To this end, PBMC cultures from healthy men were treated with DCM (15-500 ppm) for 72 h. Cell cultures were harvested and processed according to classical cytogenetic techniques. The frequency of sister chromatid exchanges (SCEs), the mitotic index (MI), the cell proliferation kinetic (CPK) value, and the level of GSTT1 activity were determined. DCM exposure decreased the MI in a dose-dependent manner in all individuals tested (20). The CPK value decreased from 125 ppm DCM, and the SCEs frequency increased from 60 ppm DCM. A significantly different response was observed when the group of individuals with low GSTT1 enzymatic activity (4 individuals), the group with medium GSTT1 activity (10 individuals), and the group of individuals with high GSTT1 enzymatic activity (6 individuals) were compared (0.077 ± 0.0124, 0.325 ± 0.0269, and 7.365 ± 1.3474 nmol HCOH/min/mg protein, respectively). These differences were reflected in the amount of change for all of the evaluated cytogenetic parameters (p<0.05, ANOVA) and indicated a clear susceptibility to DCM genotoxic effects related to GSTT1 activity because the cytogenetic effects were directly related to the GSTT1-specific activity. DCM was highly cytotoxic in PBMCs, even at doses within the safety range. Due to this toxicity, a review of the maximal limits for occupational exposure to DCM is advised.
Article
Purpose Majority of the gallbladder cancer (GBC) cases are diagnosed at an advanced stage where chemotherapy alone (or in combination with other treatment methods) is mainly opted as therapeutic approach. However, success or failure of this approach largely depends on the interindividual genetic differences. Careful consideration on the genetic association could assist in the evaluation of patient’s treatment response and survival rate. Hence, the present study aims to investigate the survival of patients with GBC and their treatment response to gemcitabine and cisplatin/carboplatin-based chemotherapy in association with Glutathione S-transferase (GSTs) gene polymorphism. Material and Methods A total of 216 histologically confirmed cases of gallbladder cancer were recruited. A total of 180 patients were treated with gemcitabine and cisplatin/carboplatin-based chemotherapy. GSTM1, GSTT1, and GSTP1 genotypes were determined by multiplex PCR and by PCR restriction fragment length polymorphism (PCR-RFLP), respectively. The influence of genetic polymorphism on overall survival was analyzed by Kaplan–Meier method, survival rate difference was analyzed by log-rank test, and hazard ratio for mortality outcomes was estimated using Cox regression method. Results GBC patients having genotype GSTP1 ( AG + GG ) showed poor 3-year survival rate of 0.8% compared to 10.9% of GSTP1 ( AA ) genotype (χ ² = 6.456, P = 0.011). The multivariate Cox regression results showed that the death risk was significantly higher in GSTP1 (AG + GG) genotype (HR = 3.858, P = 0.050). We found no association of GSTM1 and GSTT1 gene polymorphism with the survival; however, the combined genotypes of GSM1/GSTP1, GSTT1/GSTP1, and GSTM1/GSTT1/GSTP1 were associated with survival ( P = 0.053, 0.006, and 0.058, respectively). Increased death hazard was noted by the genotype combinations of GSTM1+/GSTP1AG + GG (HR = 3.484, P = 0.024), GSTM1-/GSTP1AG + GG (HR = 2.721, P = 0.014), GSTT1+/GSTP1AG + GG (HR = 20.690, P = 0.001), and GSTT1-/GSTP1AA (HR = 26.111, P < 0.0001). Our findings indicate that chemotherapy treatment response of GSTP1 (AG + GG ) has 1.62-fold increased risk for progression compared to GSTP1 (AA ) genotype (p = 0.018); however, none of the genotypes showed association with overall survival and death risk after chemotherapeutic treatment. Conclusion We found that the presence of GSTP1 ( AG + GG) genotype showed survival disadvantage and poor treatment outcomes in response to gemcitabine and cisplatin/carboplatin-based chemotherapy. This could serve as biomarker, and future research in pharmacogenomics will definitely pave the way for the development of better treatment approach for GBC.
Article
Résumé La survenue de diabètes induits ou aggravés par les corticoïdes est une situation clinique fréquente qui nécessite parfois un traitement urgent comme l'initiation de l'insulinothérapie. Les corticoïdes induisent une insulinorésistance au niveau du foie, de l'adipocyte et du muscle squelettique ainsi qu'une altération directe de la sécrétion d'insuline. Le développement d'une insulinorésistance et l'absence d'augmentation de l'insulinosécrétion sont des facteurs clefs dans la physiopathologie du diabète induit ou aggravé par les corticoïdes. Parce que le défaut d'adaptabilité de la cellule β pancréatique est en grande partie génétiquement déterminé, les antécédents familiaux de diabète (en particulier de type 2) constituent un facteur de risque important de diabète cortico-induit. Le plus souvent, la corticothérapie est utilisée à forte dose puis diminuée jusqu'à la dose minimale capable de contrôler la pathologie. Le risque de dysglycémie et son intensité vont varier en fonction de la dose et de la durée de la corticothérapie lors de ces ajustements thérapeutiques. Une prise en charge médicamenteuse du diabète est nécessaire en particulier lorsque les glycémies dépassent ≥ 2,16 g/l (12 mmol/l) à plusieurs reprises. L'insulinothérapie est le plus souvent nécessaire lorsque les glycémies dépassent 3,6 g/l (20 mmol/l) avec des symptômes cliniques. Celle-ci pourra néanmoins être relayée ultérieurement par des antidiabétiques oraux lors de la décroissance parallèle des glycémies et de la dose de corticoïdes. Le patient sera éduqué de façon similaire à toute autre forme de diabète en particulier concernant le risque d'hypoglycémie lors de la décroissance et du sevrage de la corticothérapie.
Chapter
Chapter 7 first discusses various mechanisms that can account for drug resistance and examples of approaches used to circumvent it. Mechanisms of resistance that are highlighted include altered drug target, overproduction of the target protein, overproduction of substrate or ligand, increased drug-destroying mechanisms, decreased prodrug activation, new pathways that circumvent drug action, reversal of drug action, and altered drug distribution to the site of action. A separate section discusses drug synergism and various approaches used to capitalize on synergism for improved therapeutic effects or enhanced safety margin. Specific topics of drug synergism include inhibition of a drug destroying enzyme, sequential blocking, inhibition of multiple pathways, efflux pump inhibition, and use of multiple drugs for the same target.