Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Biomolecules ; 13(4)2023 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-37189361

RESUMO

Multidrug resistance is a significant barrier that makes anticancer therapies less effective. Glutathione transferases (GSTs) are involved in multidrug resistance mechanisms and play a significant part in the metabolism of alkylating anticancer drugs. The purpose of this study was to screen and select a lead compound with high inhibitory potency against the isoenzyme GSTP1-1 from Mus musculus (MmGSTP1-1). The lead compound was selected following the screening of a library of currently approved and registered pesticides that belong to different chemical classes. The results showed that the fungicide iprodione [3-(3,5-dichlorophenyl)-2,4-dioxo-N-propan-2-ylimidazolidine-1-carboxamide] exhibited the highest inhibition potency (ΙC50 = 11.3 ± 0.5 µΜ) towards MmGSTP1-1. Kinetics analysis revealed that iprodione functions as a mixed-type inhibitor towards glutathione (GSH) and non-competitive inhibitor towards 1-chloro-2,4-dinitrobenzene (CDNB). X-ray crystallography was used to determine the crystal structure of MmGSTP1-1 at 1.28 Å resolution as a complex with S-(p-nitrobenzyl)glutathione (Nb-GSH). The crystal structure was used to map the ligand-binding site of MmGSTP1-1 and to provide structural data of the interaction of the enzyme with iprodione using molecular docking. The results of this study shed light on the inhibition mechanism of MmGSTP1-1 and provide a new compound as a potential lead structure for future drug/inhibitor development.


Assuntos
Glutationa S-Transferase pi , Glutationa Transferase , Animais , Camundongos , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/metabolismo , Simulação de Acoplamento Molecular , Glutationa Transferase/metabolismo , Glutationa/metabolismo , Isoenzimas/metabolismo , Cinética
2.
Talanta ; 251: 123796, 2023 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-35952503

RESUMO

Pi-class glutathione S-transferase (GSTP1) is a detoxification enzyme that is highly expressed in various types of cancer cells and is a promising target for cancer imaging and therapy. Ps-TAc, an acetylated derivative of the GSTP1-specific fluorogenic substrate Ps-TG, is attracting attention as an effective GSTP1 fluorescent probe, and has been successfully used to visualize intracellular GSTP1 activity. Ps-TAc is a prodrug type fluorescent probe in which the phenolic hydroxyl group of Ps-TG is acetylated and thus is susceptible to nonspecific hydrolysis, potentially compromising its ability to detect GSTP1 activity. Here, we describe the development of a highly selective fluorogenic GSTP1 substrate that is membrane permeable and does not involve esterification and show its application to live-cell imaging and FACS analysis. We designed and synthesized several compounds with benzylsulfone substituents instead of the mesyl group of Ps-TG and tested their fluorescence activation by GSTP1 catalysis in vitro and in cellulo. Of the test compounds, Ps-TG3 was the most suitable for the visualization of intracellular GSTP1 activity because the signal from living cells increased significantly when MK-571, an inhibitor of multidrug resistance proteins (MRPs), was simultaneously loaded. The results obtained by co-loading Ps-TG3 and MK571 into GSTP1-nonexpressing cells suggest that Ps-TG3 can be a substrate for MRPs. The usefulness of Ps-TG3 was demonstrated by fluorescence imaging of several cancer cell cultures and FACS analysis of lymphoma cells. The results presented here suggest that Ps-TG3, in combination with MK571, is useful for visualizing and detecting intracellular GSTP1 activity in cancer cells that highly express GSTP1.


Assuntos
Neoplasias , Pró-Fármacos , Subfamília B de Transportador de Cassetes de Ligação de ATP , Corantes Fluorescentes/química , Glutationa/química , Glutationa S-Transferase pi/química , Glutationa Transferase/química , Humanos , Pró-Fármacos/farmacologia
3.
Molecules ; 26(19)2021 Sep 24.
Artigo em Inglês | MEDLINE | ID: mdl-34641326

RESUMO

Nitric oxide is a diatomic gas that has traditionally been viewed, particularly in the context of chemical fields, as a toxic, pungent gas that is the product of ammonia oxidation. However, nitric oxide has been associated with many biological roles including cell signaling, macrophage cytotoxicity, and vasodilation. More recently, a model for nitric oxide trafficking has been proposed where nitric oxide is regulated in the form of dinitrosyl-dithiol-iron-complexes, which are much less toxic and have a significantly greater half-life than free nitric oxide. Our laboratory has previously examined this hypothesis in tumor cells and has demonstrated that dinitrosyl-dithiol-iron-complexes are transported and stored by multi-drug resistance-related protein 1 and glutathione-S-transferase P1. A crystal structure of a dinitrosyl-dithiol-iron complex with glutathione-S-transferase P1 has been solved that demonstrates that a tyrosine residue in glutathione-S-transferase P1 is responsible for binding dinitrosyl-dithiol-iron-complexes. Considering the roles of nitric oxide in vasodilation and many other processes, a physiological model of nitric oxide transport and storage would be valuable in understanding nitric oxide physiology and pathophysiology.


Assuntos
Glutationa S-Transferase pi/metabolismo , Proteínas Associadas à Resistência a Múltiplos Medicamentos/metabolismo , Neoplasias/metabolismo , Óxido Nítrico/metabolismo , Sítios de Ligação , Transporte Biológico , Regulação Neoplásica da Expressão Gênica , Glutationa S-Transferase pi/química , Humanos , Transdução de Sinais
4.
Int J Mol Sci ; 22(8)2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33920860

RESUMO

Glutathione transferases (GSTs) form a family of detoxication enzymes instrumental in the inactivation and elimination of electrophilic mutagenic and carcinogenic compounds. The Pi class GST P1-1 is present in most tissues and is commonly overexpressed in neoplastic cells. GST P1-1 in the dog, Canis lupus familiaris, has merits as a marker for tumors and as a target for enzyme-activated prodrugs. We produced the canine enzyme CluGST P1-1 by heterologous bacterial expression and verified its cross-reactivity with antihuman-GST P1-1 antibodies. The catalytic activity with alternative substrates of biological significance was determined, and the most active substrate found was benzyl isothiocyanate. Among established GST inhibitors, Cibacron Blue showed positive cooperativity with an IC50 value of 43 nM. Dog GST P1-1 catalyzes activation of the prodrug Telcyta, but the activity is significantly lower than that of the human homolog.


Assuntos
Glutationa S-Transferase pi/metabolismo , Medicina Veterinária , Sequência de Aminoácidos , Animais , Biocatálise , Cães , Glutationa S-Transferase pi/antagonistas & inibidores , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/isolamento & purificação , Humanos , Modelos Moleculares , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Estrutura Terciária de Proteína , Especificidade por Substrato
5.
J Med Chem ; 63(17): 9965-9976, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32787104

RESUMO

Synthetic triterpenoids including CDDO, its methyl ester (CDDO-Me, bardoxolone methyl), and its imidazolide (CDDO-Im) enhance Nrf2-mediated antioxidant and anti-inflammatory activity in many diseases by reacting with thiols on the adaptor protein, Keap1. Unlike monofunctional CDDO-Me, the bifunctional analog, CDDO-Im, has a second reactive site (imidazolide) and can covalently bind to amino acids other than cysteine on target proteins such as glutathione S-transferase pi (GSTP), serum albumin, or Keap1. Here we show for the first time that bifunctional CDDO-Im (in contrast to CDDO-Me), as low as 50 nM, can covalently transacylate arginine and serine residues in GSTP and cross-link them to adjacent cysteine residues. Moreover, we show that CDDO-Im binds covalently to Keap1 by forming permanent Michael adducts with eight different cysteines, and acyl adducts with lysine and several tyrosine residues. Modeling studies suggest that the Tyr 85 adduct stabilizes the Keap1-Cul3 complex, thereby enhancing the potency of CDDO-Im.


Assuntos
Imidazóis/química , Proteína 1 Associada a ECH Semelhante a Kelch/química , Ácido Oleanólico/análogos & derivados , Sequência de Aminoácidos , Proteínas Culina/química , Proteínas Culina/metabolismo , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/metabolismo , Humanos , Imidazóis/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Simulação de Acoplamento Molecular , Ácido Oleanólico/química , Ácido Oleanólico/metabolismo , Multimerização Proteica/efeitos dos fármacos , Albumina Sérica Humana/química , Albumina Sérica Humana/metabolismo
6.
Fish Shellfish Immunol ; 105: 144-151, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32652299

RESUMO

Glutathione S-transferases (GSTs) play important roles in immunity by protecting organisms against the damage of reactive oxygen species (ROS). In this study, a pi-class GST cDNA sequence was first cloned from noble scallop Chlamys nobilis (named CnGSTp). The full length cDNA of CnGSTp was 922 bp, encoding a cytosolic protein of 202 amino acids residues, with predicted molecular masses of 23.1 kDa. Then an acute Vibrio Parahaemolyticus challenge experiment was conducted by using the Golden and Brown noble scallops with different total carotenoids content (TCC), and CnGSTp expression level, TCC and ROS level was separately determined. The results showed that ROS and CnGSTp expression levels were significantly up-regulate under Vibrio Parahaemolyticus challenge than the control group (P < 0.05). The Golden scallops showed significantly higher CnGSTp expression level and lower ROS level in hemocytes than the Brown ones (P < 0.05). Moreover, there is a significantly positive correlation between TCC and ROS in the Golden scallops. The present results revealed that CnGSTp plays important roles in immune response and carotenoids play assistant roles in antioxidant defense system under pathogenic stress in the noble scallop.


Assuntos
Regulação da Expressão Gênica/imunologia , Glutationa S-Transferase pi/genética , Glutationa S-Transferase pi/imunologia , Imunidade Inata/genética , Pectinidae/genética , Pectinidae/imunologia , Sequência de Aminoácidos , Animais , Antioxidantes/metabolismo , Sequência de Bases , Expressão Gênica , Perfilação da Expressão Gênica , Glutationa S-Transferase pi/química , Pectinidae/enzimologia , Filogenia , Alinhamento de Sequência
7.
Sci Rep ; 10(1): 6032, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32265484

RESUMO

The Glutathione S-transferases (GSTs) protects cellular DNA against oxidative damage. The role of GSTP1 polymorphism (A313G; Ile105Val) as a susceptibility factor in oral cancer was evaluated in a hospital-based case-control study in North-East India, because the habit of chewing raw areca-nut (RAN) with/without tobacco is common in this region. Genetic polymorphism was investigated by genotyping 445 cases and 444 controls. Individuals with the GSTP1 AA-genotype showed association with the oral cancer (OR = 3.1, 95% CI = 2.4-4.2, p = 0.0002). Even after adjusting for age, sex and habit the AA-genotype is found to be significantly associated with oral cancer (OR = 2.4, 95% CI = 1.7-3.2, p = 0.0001). A protein-protein docking analysis demonstrated that in the GG-genotype the binding geometry between c-Jun Kinase and GSTP1 was disrupted. It was validated by immunohistochemistry in human samples, showing lower c-Jun-phosphorylation and down-regulation of pro-apoptotic genes in normal oral epithelial cells with the AA-genotype. In silico docking revealed that AA-genotype weakly detoxifies the RAN/tobacco metabolites. In addition, experiments revealed a higher level of 8-Oxo-2'-deoxyguanosine induction in tumor samples with the AA-genotype. Thus, habit of using RAN/tobacco and GSTP1 AA-genotype together play a significant role in predisposition to oral cancer risk by showing higher DNA-lesions and lower c-Jun phosphorylation that may inhibit apoptosis.


Assuntos
Glutationa S-Transferase pi/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Bucais/genética , Neoplasias Bucais/metabolismo , Polimorfismo de Nucleotídeo Único , Adulto , Idoso , Idoso de 80 Anos ou mais , Alelos , Areca/metabolismo , Cristalografia por Raios X , Dano ao DNA , Feminino , Predisposição Genética para Doença , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Neoplasias Bucais/etiologia , Estresse Oxidativo , Fosforilação , Uso de Tabaco/efeitos adversos , Uso de Tabaco/metabolismo
8.
Chem Biol Drug Des ; 94(6): 2094-2102, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31452310

RESUMO

Quantum dots (QD) are being evaluated as inorganic nanoparticles for both in vitro and in vivo optical imaging. They are also used as sensors or vehicles for targeted drug delivery combined with optical imaging. In this study, we demonstrated that glutathione-coated Ag2 S QDs (GSH-Ag2 S QDs) act as a substrate analogue of glutathione S-transferase (GST) enzymes for the first time in the literature. The GSTs belong to a major group of detoxification enzymes involved in the detoxification metabolism responsible for the protection of cells against reactive oxygen species (ROS) or electrophiles. GST isozymes are impaired in the various diseases such as neurological diseases and cancer. We evaluated the interaction of GST-pi enzyme with GSH-Ag2 S QDs, which have never been studied in the literature before, using both fluorometric and spectrophotometric methods. Our data showed that GSH-Ag2 S QDs gave reaction with GST enzyme as a substrate analogue. In conclusion, our data may help to guide researchers for further development of sensing systems for GST activity which is impaired in various diseases including cancer.


Assuntos
Glutationa S-Transferase pi/metabolismo , Glutationa/química , Pontos Quânticos/química , Compostos de Prata/química , Animais , Linhagem Celular Tumoral , Glutationa S-Transferase pi/química , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Cinética , Fígado/enzimologia , Fígado/metabolismo , Pontos Quânticos/metabolismo , Ratos , Ratos Wistar , Especificidade por Substrato
9.
Talanta ; 204: 633-640, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31357346

RESUMO

Fluorogenic substrates are used to visualize the activity of cancer-associated enzymes and to interpret biological events. Certain types of glutathione S-transferase (GST), such as Pi class GST (referred to as GSTP1), are more highly expressed in a wide variety of human cancer tissues compared to their corresponding normal tissues. Pi class GST is thus a cancer cell molecular marker and potential target for overcoming resistance to chemotherapy. Here, we report that 4-bromo-1,8-naphthalimide (BrNaph) is a practical fluorogenic GST substrate. We have found that HE-BrNaph, an N-hydroxyethyl derivative, shows remarkable fluorescence enhancement upon GST-catalyzed SNAr replacement of the bromo group with a glutathionyl group. This substitution was highly selective and occurred only in the presence of GSH/GSTs; no non-enzymatic reaction was observed. We demonstrated that HE-BrNaph allows visualization of GST activity in living cells and enables to distinguish cancer cells from normal cells. Further, various N-substitutions in BrNaph retain susceptibility to enzymatic activity and isozyme selectivity, suggesting the applicability of BrNaph derivatives. Thus, BrNaph and its derivatives are GST substrates useful for fluorescence imaging and the intracellular detection of GSTP1 activity in living cells.


Assuntos
Corantes Fluorescentes/química , Glutationa S-Transferase pi/análise , Naftalimidas/química , Linhagem Celular Tumoral , Ensaios Enzimáticos/métodos , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/toxicidade , Glutationa S-Transferase pi/química , Humanos , Cinética , Microscopia Confocal/métodos , Microscopia de Fluorescência/métodos , Naftalimidas/síntese química , Naftalimidas/toxicidade , Neoplasias/diagnóstico
10.
Proc Natl Acad Sci U S A ; 116(28): 13943-13951, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31221747

RESUMO

Cisplatin [cis-diamminedichloroplatinum(II) (cis-DDP)] is one of the most successful anticancer agents effective against a wide range of solid tumors. However, its use is restricted by side effects and/or by intrinsic or acquired drug resistance. Here, we probed the role of glutathione transferase (GST) P1-1, an antiapoptotic protein often overexpressed in drug-resistant tumors, as a cis-DDP-binding protein. Our results show that cis-DDP is not a substrate for the glutathione (GSH) transferase activity of GST P1-1. Instead, GST P1-1 sequesters and inactivates cisplatin with the aid of 2 solvent-accessible cysteines, resulting in protein subunits cross-linking, while maintaining its GSH-conjugation activity. Furthermore, it is well known that GST P1-1 binding to the c-Jun N-terminal kinase (JNK) inhibits JNK phosphorylation, which is required for downstream apoptosis signaling. Thus, in turn, GST P1-1 overexpression and Pt-induced subunit cross-linking could modulate JNK apoptotic signaling, further confirming the role of GST P1-1 as an antiapoptotic protein.


Assuntos
Cisplatino/química , Glutationa S-Transferase pi/química , Proteínas Quinases JNK Ativadas por Mitógeno/química , Neoplasias/tratamento farmacológico , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glutationa/química , Glutationa S-Transferase pi/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Neoplasias/genética , Fosforilação , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Transdução de Sinais/efeitos dos fármacos
11.
Chembiochem ; 20(7): 900-905, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30548113

RESUMO

Glutathione S-transferase π (GSTP1-1 ) is overexpressed in many types of cancer and is involved in drug resistance. Therefore, GSTP1-1 is an important target in cancer therapy, and many GST inhibitors have been reported. We had previously developed an irreversible inhibitor, GS-ESF, as an effective GST inhibitor; however, its cellular permeability was too low for it to be used in inhibiting intracellular GST. We have now developed new irreversible inhibitors by introducing sulfonyl fluoride (SF) into chloronitrobenzene (CNB). The mechanism of action was revealed to be that CNBSF first reacts with glutathione (GSH) through an aromatic substitution in the cell, then the sulfonyl group on the GSH conjugate with CNBSF reacts with Tyr108 of GST to form a sulfonyl ester bond. Our new inhibitor irreversible inhibited GSTP1-1 both in vitro and in cellulo with a long duration of action.


Assuntos
Inibidores Enzimáticos/farmacologia , Glutationa S-Transferase pi/antagonistas & inibidores , Glutationa/análogos & derivados , Glutationa/farmacologia , Sulfonas/farmacologia , Sequência de Aminoácidos , Sítios de Ligação/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/síntese química , Glutationa/síntese química , Glutationa S-Transferase pi/química , Humanos , Simulação de Acoplamento Molecular , Sulfonas/síntese química , Tirosina/química
12.
Free Radic Biol Med ; 129: 486-491, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30315934

RESUMO

The development of high-throughput technologies in the last decade produced an exponential increase in the amount of biological data available. The case of redox biology and apoptosis is not an exception, and nowadays there is a need to integrate information from multiple "omics" studies. Therefore, validation of proposed discoveries is essential. However, the study in biological systems of the effect of the massive amounts of sequence variation data generated with next-generation sequencing (NGS) technologies can be a very difficult and expensive process. In this context, the present study aimed to demonstrate the advantages of a computational methodology to systematically analyze the structural and functional effects of protein variants, in order to prioritize further studies. This approach stands out for its easy implementation, low costs and low time consumed. First, the possible impact of mutations on protein structure and function was tested by a combination of tools based on evolutionary and structural information. Next, homology modeling was performed to predict and compare the 3D protein structures of unresolved amino acid sequences obtained from genomic resequencing. This analysis applied to the bovine GSTP1 allowed to determine that some of amino acid substitutions may generate important changes in protein structure and function. Moreover, the haplotype analysis highlighted three structure variants worthwhile studying through in vitro or in vivo experiments.


Assuntos
Substituição de Aminoácidos , Glutationa S-Transferase pi/química , Mutação , Polimorfismo de Nucleotídeo Único , RNA Mensageiro/genética , Sequência de Aminoácidos , Animais , Bovinos , Biologia Computacional/métodos , Expressão Gênica , Glutationa S-Transferase pi/genética , Glutationa S-Transferase pi/metabolismo , Haplótipos , Conformação Proteica , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Homologia Estrutural de Proteína , Relação Estrutura-Atividade
13.
Talanta ; 179: 845-852, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-29310316

RESUMO

Pi class glutathione S-transferase (GSTP1) is highly expressed in various cancerous cells and pre-neoplastic legions, where it is involved in apoptotic resistance or metabolism of several anti-tumour chemotherapeutics. Therefore, GSTP1 is a marker of malignant and pre-malignant cells and is a promising target for visualization and drug development. Here we demonstrate that fluorescein diacetate (FDA), a fluorescent probe used for vital staining, is a fluorescently activated by esterolytic activity of human GSTP1 (hGSTP1) selectively among various cytosolic GSTs. Fluorescence activation of FDA susceptible to GST inhibitors was observed in MCF7 cells exogenously overexpressing hGSTP1, but not in cells overexpressing hGSTA1 or hGSTM1. Inhibitor-sensitive fluorescence activation was also observed in several cancer cell lines endogenously expressing GSTP1, suggesting that GSTP1 is involved in FDA esterolysis in these cells. Among the FDA derivatives examined, FOMe-Ac, the acetyl ester of fluorescein O-methyl ether, was found to be a potential reporter for GSH-dependent GSTP1 activity as well as for carboxylesterase activity. Since GSTP1 is highly expressed in various types of cancer cells compared to their normal counterparts, improving the fluorogenic substrates to be more selective to the esterolysis activity of GSTP1 rather than carboxylesterases should lead to development of tools for detecting GSTP1-overexpressing cancer cells and investigating the biological functions of GSTP1.


Assuntos
Biomarcadores Tumorais/química , Fluoresceínas/química , Corantes Fluorescentes/química , Glutationa S-Transferase pi/química , Biomarcadores Tumorais/antagonistas & inibidores , Glutationa/química , Glutationa S-Transferase pi/antagonistas & inibidores , Glutationa Transferase/química , Células HeLa , Humanos , Células MCF-7 , Oxidiazóis/química , Espectrometria de Fluorescência , Especificidade por Substrato
14.
Biosci Rep ; 38(1)2018 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-29358310

RESUMO

The 7-nitro-2,1,3-nitrobenzoxadiazole (NBD) derivatives are a series of compounds containing the NBD scaffold that are not glutathione (GSH) peptidomimetics, and result in a strong inhibition of glutathione S-transferases (GSTs). Growing evidences highlight their pivotal roles and outstanding anticancer activity in different tumor models. In particular, 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio) hexanol (NBDHEX) is extensively studied, which is a very efficient inhibitor of GSTP1-1. It triggers apoptosis in several tumor cell lines and this cytotoxic activity is observed at micro and submicromolar concentrations. Importantly, studies have shown that NBDHEX acts as an anticancer drug by inhibiting GSTs catalytic activity, avoiding inconvenience of the inhibitor extrusion from the cell by specific pumps and disrupting the interaction between the GSTP1-1 and key signaling effectors. Additionally, some researchers also have discovered that NBDHEX can act as late-phase autophagy inhibitor, which opens new opportunities to fully exploit its therapeutic potential. In this review, we summarize the advantages, anticancer mechanisms, and analogs of this compound, which will establish the basis on the usage of NBDHEX in clinical applications in future.


Assuntos
Antineoplásicos/química , Glutationa S-Transferase pi/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Oxidiazóis/química , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Azóis/química , Azóis/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glutationa S-Transferase pi/química , Hexanóis/química , Hexanóis/uso terapêutico , Humanos , Neoplasias/patologia , Nitrobenzenos/química , Nitrobenzenos/uso terapêutico , Oxidiazóis/uso terapêutico
15.
Artif Cells Nanomed Biotechnol ; 46(3): 510-517, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28503938

RESUMO

The glutathione transferases (GSTs) are a family of widely distributed Phase II detoxification enzymes. GST P1-1 is frequently overexpressed in rat and human tumours. It is suggested that overexpression of hGST P1-1 by human tumor cells may play a role in resistance to cancer chemotherapy. Hence, hGST P1-1 can be a promising target for cancer treatment. In this study, new hGST P1-1 inhibitors, 2-(4-substitutedphenyl/benzyl)-5-(4-trifluoromethylphenylsulphonamido) benzoxazole derivatives (Va-Vk) have been designed and synthesized. Surprisingly, in vitro hGST P1-1 enzyme inhibition studies demonstrated that all of the tested compounds except Vj had better activity than the reference drug EA and it is also correlated with the docking results. Additionally we compared the interactions with hGST P1-1 enzyme of newly synthesized compound Vh (bearing CF3 group) and previously synthesized compound 5f (bearing NO2 group). According to the docking results, compound Vh bound to the hGST P1-1 enzyme with a higher affinity compared to 5f. Therefore, we can consider that these data make a sense and can explain its higher activity. The compounds that obtained from this research could be used as scaffolds in design of new potent hGST P1-1 inhibitors useful in the treatment of the resistance of cancer chemotherapy.


Assuntos
Benzoxazóis , Inibidores Enzimáticos , Glutationa S-Transferase pi , Simulação de Acoplamento Molecular , Benzoxazóis/síntese química , Benzoxazóis/química , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Glutationa S-Transferase pi/antagonistas & inibidores , Glutationa S-Transferase pi/química , Humanos
16.
J Biochem Mol Toxicol ; 31(10)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28621814

RESUMO

Zebrafish has in recent years emerged as a popular vertebrate model for use in pharmacological and toxicological studies. While there have been sporadic studies on the zebrafish glutathione S-transferases (GSTs), the zebrafish GST gene superfamily still awaits to be fully elucidated. We report here the identification of 15 zebrafish cytosolic GST genes in NCBI GenBank database and the expression, purification, and enzymatic characterization of the zebrafish cytosolic GST Pi-1 (GSTP1). The cDNA encoding the zebrafish GSTP1 was cloned from a 3-month-old female zebrafish, expressed in Eschelichia coli host cells, and purified. Purified GSTP1 displayed glutathione-conjugating activity toward 1-chloro-2,4-dinitrobenzene as a representative substrate. The enzymatic characteristics of the zebrafish GSTP1, including pH-dependency, effects of metal cations, and kinetic parameters, were studied. Moreover, the expression of zebrafish GSTP1 at different developmental stages during embryogenesis, throughout larval development, onto maturity was examined.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Glutationa S-Transferase pi , Proteínas de Peixe-Zebra , Peixe-Zebra/metabolismo , Animais , Dinitroclorobenzeno/química , Feminino , Glutationa S-Transferase pi/biossíntese , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/genética , Especificidade por Substrato , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/química , Proteínas de Peixe-Zebra/genética
17.
FEBS J ; 284(15): 2442-2463, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28622459

RESUMO

The glutathione transferase (GST) gene family is divided into 14 classes in photosynthetic organisms. Among them, the Phi class (GSTF) is composed of a large number of genes that are often induced in response to environmental constraints due to their ability to detoxify xenobiotics, to their peroxidase activity and to their involvement in the biosynthesis and/or transport of secondary metabolites. However, the exact functions of GSTFs from many plants including Populus trichocarpa are unknown. Here, following GSTF1 characterization, we have performed a comparative analysis of the seven other GSTFs found in poplar by systematically evaluating the biochemical and enzymatic properties of the corresponding recombinant proteins and of variants mutated for active site residues and by determining the three-dimensional structures of several representatives. Owing to the presence of a cysteine with a pKa value around 5 in their active site, GSTF3, F7, and F8 displayed a thiol transferase activity in addition to the usual glutathione transferase and peroxidase activities. From structural analyses, it appeared that these dual biochemical properties originate from the existence of a certain variability in the ß1-α1 loop. This allows positioning of several active site residues at proximity of the glutathione molecule, which itself remains unchanged in GSTF three-dimensional structures. These results highlight the promiscuity of some GSTFs and that changes of active site residues in some isoforms during evolution generated functional diversity by modifying their activity profile. DATABASE: Structural data are available in the PDB under the accession numbers 5EY6, 5F05, 5F06, and 5F07.


Assuntos
Glutationa S-Transferase pi/metabolismo , Modelos Moleculares , Proteínas de Plantas/metabolismo , Populus/enzimologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Substituição de Aminoácidos , Biocatálise , Domínio Catalítico , Cisteína/química , Dimerização , Estabilidade Enzimática , Glutationa/química , Glutationa/metabolismo , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/genética , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Mutagênese Sítio-Dirigida , Mutação , Filogenia , Proteínas de Plantas/química , Proteínas de Plantas/genética , Conformação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência
18.
Gene ; 609: 19-27, 2017 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-28153749

RESUMO

Gene polymorphisms lead to varied structure and functional properties. A single nucleotide polymorphism (SNP) i.e. Ile105Val (rs1695) in glutathione S-transferase P1 (GSTP1) gene influences cytological toxicity and modulates the risk to occupational diseases. Apart from this, cancer, neuropathy, NOx, SOx and ozone mediated respiratory function decline including lung inflammation, asthma, allergy etc., have been reported in people with this missense mutation. Here, the functional properties of rs1695 polymorph are revisited through a computational approach. Changes incurred by GSTP1 antioxidant protein as a result of alteration in its sequence, have been studied through docking followed by Poisson-Boltzmann electrostatic equation interpretation, grid and coulombic energy profile mapping for protein polymorphs with DelPhi. Molecular docking simulation of variant and wild type (WT) protein was carried out with eight FDA approved compounds that target GSTP1 for treatment of various diseases. This was to observe binding pattern variation upon mutation induction. Grid, reaction field and coulombic energy calculation of WT and mutated polymorph, complexed with and without these moieties was then attempted. Alteration in conformation and energy was observed in apo- and holo- form of GSTP1 and their ligand-bound complexes as a result of this mutation. This study is a demo of appraising gene-environment interaction based deleteriousness through molecular docking and dynamics simulation approach.


Assuntos
Glutationa S-Transferase pi/química , Substituição de Aminoácidos , Metabolismo Energético , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Polimorfismo de Nucleotídeo Único
19.
J Biol Chem ; 292(1): 112-120, 2017 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-27872191

RESUMO

Glutathione S-transferase pi 1 (GSTP1) is frequently overexpressed in cancerous tumors and is a putative target of the plant compound piperlongumine (PL), which contains two reactive olefins and inhibits proliferation in cancer cells but not normal cells. PL exposure of cancer cells results in increased reactive oxygen species and decreased GSH. These data in tandem with other information led to the conclusion that PL inhibits GSTP1, which forms covalent bonds between GSH and various electrophilic compounds, through covalent adduct formation at the C7-C8 olefin of PL, whereas the C2-C3 olefin of PL was postulated to react with GSH. However, direct evidence for this mechanism has been lacking. To investigate, we solved the X-ray crystal structure of GSTP1 bound to PL and GSH at 1.1 Å resolution to rationalize previously reported structure activity relationship studies. Surprisingly, the structure showed that a hydrolysis product of PL (hPL) was conjugated to glutathione at the C7-C8 olefin, and this complex was bound to the active site of GSTP1; no covalent bond formation between hPL and GSTP1 was observed. Mass spectrometry (MS) analysis of the reactions between PL and GSTP1 confirmed that PL does not label GSTP1. Moreover, MS data also indicated that nucleophilic attack on PL at the C2-C3 olefin led to PL hydrolysis. Although hPL inhibits GSTP1 enzymatic activity in vitro, treatment of cells susceptible to PL with hPL did not have significant anti-proliferative effects, suggesting that hPL is not membrane-permeable. Altogether, our data suggest a model wherein PL is a prodrug whose intracellular hydrolysis initiates the formation of the hPL-GSH conjugate, which blocks the active site of and inhibits GSTP1 and thereby cancer cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Dioxolanos/farmacologia , Glutationa S-Transferase pi/química , Glutationa S-Transferase pi/metabolismo , Glutationa/metabolismo , Neoplasias Pancreáticas/patologia , Cristalografia por Raios X , Humanos , Espectrometria de Massas , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/enzimologia , Ligação Proteica , Conformação Proteica , Células Tumorais Cultivadas
20.
Protein Sci ; 26(2): 317-326, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27863446

RESUMO

Arsenic-based compounds are paradoxically both poisons and drugs. Glutathione transferase (GSTP1-1) is a major factor in resistance to such drugs. Here we describe using crystallography, X-ray absorption spectroscopy, mutagenesis, mass spectrometry, and kinetic studies how GSTP1-1 recognizes the drug phenylarsine oxide (PAO). In conditions of cellular stress where glutathione (GSH) levels are low, PAO crosslinks C47 to C101 of the opposing monomer, a distance of 19.9 Å, and causes a dramatic widening of the dimer interface by approximately 10 Å. The GSH conjugate of PAO, which forms rapidly in cancerous cells, is a potent inhibitor (Ki = 90 nM) and binds as a di-GSH complex in the active site forming part of a continuous network of interactions from one active site to the other. In summary, GSTP1-1 can detoxify arsenic-based drugs by sequestration at the active site and at the dimer interface, in situations where there is a plentiful supply of GSH, and at the reactive cysteines in conditions of low GSH.


Assuntos
Arsênio/química , Arsenicais/química , Reagentes de Ligações Cruzadas/química , Glutationa S-Transferase pi/química , Humanos , Multimerização Proteica , Estrutura Quaternária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA