Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 975
1.
Biochemistry (Mosc) ; 89(3): 553-561, 2024 Mar.
Article En | MEDLINE | ID: mdl-38648772

Glutathione S-transferases (GSTs) belong to the superfamily of multifunctional detoxification isoenzymes with an important role in cellular signaling. They can prevent reactive electrophilic compounds from harming the body by covalently binding identical type of moleculs to each other. GSTs can be used alone or in combination for cancer detection or diagnosis, in addition to therapeutic interventions. In recent years, indoles have become important due to their structural properties and biological activities such as antitubercular, antiulcer, anti-oxidant, and antidiabetic, as well as for the development of new anticancer agents. The current research investigated effects of some indoles with 3-carboxaldehyde structure on the GST enzyme activity. Impacts of various concentrations of indoles on the in vitro GST activity were examined. While IC50 values for the compounds ranged from 0.042 to 1.570 mM, Ki values changed between 0.018 ± 0.01 and 1.110 ± 0.15 mM. 6-Methylindole-3-carboxaldehyde (1b) exhibited the highest inhibitory effect among the indoles examined. Indole derivatives used in the study can be evaluated in further pharmacological studies due to their effects on GST activity.


Glutathione Transferase , Indoles , Indoles/pharmacology , Indoles/chemistry , Glutathione Transferase/metabolism , Glutathione Transferase/antagonists & inhibitors , Humans , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry , Kinetics
2.
Chem Biodivers ; 20(4): e202300049, 2023 Apr.
Article En | MEDLINE | ID: mdl-36866854

Resinous beehive product propolis has many biological activities. It contains various aromatic substances that have great differences in their chemical composition depending on the natural flora. Thus, chemical characterization and biological properties of propolis samples is an important subject for the pharmaceutical industry. In this study, the propolis samples collected from three cities in Turkey were prepared as methanol (MEP), ethanol (EEP), chloroform (ChlEP), hexane (HxEP), and ethyl acetate (EAEP) extracts using an ultrasonic assisted technique. The antioxidant capacities of the samples were evaluated by free radical scavenging activity (DPPH), cation radical scavenging activity (ABTS), and reducing activity (CUPRAC) and (FRAP). The strongest biological activities were detected in ethanol and methanol extracts. Enzyme inhibition of the propolis samples were determined against the human glutathione S-transferase (GST) and angiotensin converting enzyme (ACE). IC50 values of MEP1, MEP2, and MEP3 samples against the ACE were found as 13.9 µg/mL, 14.8 µg/mL, and 12.8 µg/mL, while against the GST IC50 values of MEP1, MEP2, and MEP3 samples were as 5.92 µg/mL, 9.49 µg/mL, and 5.72 µg/mL. To know the possible causes of the biological test results advanced LC/MS/MS method was applied. trans-ferulic acid, kaempferol, and chrysin were found as the most abundant phenolic compounds in each sample. The propolis extracts obtained using the proper solvent have a good potential to be used in pharmaceuticals to treat the diseases related to oxidative damage, hypertension, and inflammation. Finally, the interactions between chrysin, trans-ferulic acid and kaempferol molecules with ACE and GST receptors were analyzed using molecular docking study. Selected molecules interact with active residues by binding to the active site of the receptors.


Antioxidants , Propolis , Humans , Angiotensins , Antioxidants/pharmacology , Antioxidants/chemistry , Ethanol , Kaempferols , Methanol/chemistry , Molecular Docking Simulation , Phenols/pharmacology , Propolis/pharmacology , Propolis/chemistry , Tandem Mass Spectrometry , Angiotensin-Converting Enzyme Inhibitors/chemistry , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/chemistry , Plant Extracts/chemistry
3.
Mol Biochem Parasitol ; 252: 111524, 2022 11.
Article En | MEDLINE | ID: mdl-36195242

Glutathione transferases (GSTs) are major detoxification enzymes vital for the survival and reproduction of schistosomes during infection in humans. Schistosoma encode two GST isoenzymes, the 26- and 28-kDa isoforms, that show different substrate specificities and cellular localisations. Bromosulfophthalein (BSP) has been identified and characterised as a potent 26-kDa Schistosoma japonicum GST (Sj26GST) inhibitor with an anthelmintic potential. This study describes the structure, function, and ligandin properties of the 28-kDa Schistosoma japonicum GST (Sj28GST) towards BSP. Enzyme kinetics show that BSP is a potent enzyme inhibitor, with a specific activity decreases from 60.4 µmol/min/mg to 0.0742 µmol/min/mg and an IC50 in the micromolar range of 0.74 µM. Far-UV circular dichroism confirmed that purified Sj28GST follows a typical GST fold, which is predominantly alpha-helical. Fluorescence spectroscopy suggests that BSP binding occurs at a site distinct from the glutathione-binding site (G-site); however, the binding does not alter the local G-site environment. Isothermal titration calorimetry studies show that the binding of BSP to Sj28GST is exergonic (∆G°= -33 kJ/mol) and enthalpically-driven, with a stoichiometry of one BSP per dimer. The stability of Sj28GST (∆G(H2O) = 4.7 kcal/mol) is notably lower than Sj26GST, owing to differences in the enzyme's dimeric interfaces. We conclude that Sj28GST shares similar biophysical characteristics with Sj26GST based on its kinetic properties and susceptibility to low concentrations of BSP. The study supports the potential benefits of re-purposing BSP as a potential drug or prodrug to mitigate the scourge of schistosomiasis.


Glutathione Transferase , Schistosoma japonicum , Sulfobromophthalein , Animals , Binding Sites , Calorimetry , Glutathione/metabolism , Glutathione Transferase/antagonists & inhibitors , Schistosoma japonicum/drug effects , Schistosoma japonicum/enzymology , Sulfobromophthalein/pharmacology
4.
Biomolecules ; 12(10)2022 09 24.
Article En | MEDLINE | ID: mdl-36291574

Glutathione transferases (GSTs) are a family of Phase II detoxification enzymes that are involved in the development of multi-drug resistance (MDR) phenomena toward chemotherapeutic agents. GST inhibitors are considered candidate compounds able to chemomodulate and reverse MDR. The natural flavonoid myricetin (MYR) has been shown to exhibit a wide range of pharmacological functions, including antitumor activity. In the present work, the interaction of MYR with human glutathione transferase A1-1 (hGSTA1-1) was investigated by kinetics inhibition analysis and molecular modeling studies. The results showed that MYR binds with high affinity to hGSTA1-1 (IC50 2.1 ± 0.2 µΜ). It functions as a non-competitive inhibitor towards the electrophile substrate 1-chloro-2,4-dinitrobenzene (CDNB) and as a competitive inhibitor towards glutathione (GSH). Chemical modification studies with the irreversible inhibitor phenethyl isothiocyanate (PEITC), in combination with in silico molecular docking studies allowed the prediction of the MYR binding site. MYR appears to bind at a distinct location, partially overlapping the GSH binding site (G-site). The results of the present study show that MYR is a potent inhibitor of hGSTA1-1 that can be further exploited towards the development of natural, safe, and effective GST-targeted cancer chemosensitizers.


Flavonoids , Glutathione Transferase , Humans , Dinitrochlorobenzene , Flavonoids/pharmacology , Glutathione/metabolism , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Kinetics , Molecular Docking Simulation
5.
Chem Commun (Camb) ; 58(24): 3917-3920, 2022 Mar 22.
Article En | MEDLINE | ID: mdl-35237781

Antioxidant-defense systems of tumor cells protect them from oxidative damage. Herein, a carrier-free nanomedicine is developed based on chlorine e6 (Ce6) and coniferyl ferulate (Con), which inhibits glutathione S-transferase (GST) activity to hamper antioxidant systems and amplify intracellular oxidative stress for enhanced photodynamic therapy.


Glutathione Transferase , Nanomedicine , Neoplasms , Photochemotherapy , Porphyrins , Glutathione Transferase/antagonists & inhibitors , Neoplasms/drug therapy , Oxidative Stress , Photosensitizing Agents/pharmacology , Porphyrins/pharmacology
6.
J Enzyme Inhib Med Chem ; 37(1): 728-742, 2022 Dec.
Article En | MEDLINE | ID: mdl-35176963

The linking of ethacrynic acid with ethylenediamine and 1,4-butanediamine gave EDEA and BDEA, respectively, as membrane-permeable divalent pro-inhibitors of glutathione S-transferase (GST). Their divalent glutathione conjugates showed subnanomolar inhibition and divalence-binding to GSTmu (GSTM) (PDB: 5HWL) at ∼0.35 min-1. In cisplatin-resistant SK-OV-3, COC1, SGC7901 and A549 cells, GSTM activities probed by 15 nM BDEA or EDEA revealed 5-fold and 1.0-fold increases in cisplatin-resistant SK-OV-3 and COC1 cells, respectively, in comparison with the susceptible parental cells. Being tolerable by HEK293 and LO2 cells, BDEA at 0.2 µM sensitised resistant SK-OV-3 and COC1 cells by ∼3- and ∼5-folds, respectively, released cytochrome c and increased apoptosis; EDEA at 1.0 µM sensitised resistant SK-OV-3 and A549 cells by ∼5- and ∼7-fold, respectively. EDEA at 1.7 µg/g sensitised resistant SK-OV-3 cells to cisplatin at 3.3 µg/g in nude mouse xenograft model. BDEA and EDEA are promising leads for probing cellular GSTM and sensitising cisplatin-resistant ovarian cancers.


Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Ethacrynic Acid/pharmacology , Ethylenediamines/pharmacology , Glutathione Transferase/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Putrescine/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cisplatin/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Screening Assays, Antitumor , Ethacrynic Acid/chemistry , Ethylenediamines/chemistry , Female , Glutathione Transferase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Mice , Mice, Nude , Molecular Structure , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Putrescine/chemistry , Structure-Activity Relationship
7.
Chem Biodivers ; 18(12): e2100433, 2021 Dec.
Article En | MEDLINE | ID: mdl-34596972

In the present study, 3-substitued-4-(4-hydroxybenzylidenamino)-4,5-dihydro-1H-1,2,4-triazol-5-ones (S1-8) were synthesized by treating 4-hydroxybenzaldehyde (B) with eight different 3-substitued-4-amino-4,5-dihydro-1H-1,2,4-triazole-5-ones (T1-8) in acetic acid medium, separately. The synthesized Schiff bases (S) were reacted with formaldehyde and secondary amine such as 4-piperidinecarboxyamide to afford novel heterocyclic bases. 3-Substitued-4-(4-hydroxybenzylidenamino)-4,5-dihydro-1H-1,2,4-triazol-5-ones (T) were treated with 4-piperidinecarboxyamide in the presence of formaldehyde to synthesize eight new 1-(4-piperidinecarboxyamide-1-yl-methyl)-3-substitued-4-(4-hydroxybenzylidenamino)-4,5-dihydro-1H-1,2,4-triazol-5-ones (M1-8). The structure characterization of compounds was carried out using 1 H-NMR, IR, HR-MS, and 13 C-NMR spectroscopic methods. The inhibitory properties of the newly synthesized compounds were calculated against the acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and glutathione S-transferase (GST) enzymes. Ki values were calculated in the range of 20.06±3.11-36.86±6.17 µM for GST, 17.87±2.91-30.53±4.25 µM for AChE, 9.08±0.69-20.02±2.88 µM for BChE, respectively, Besides, IC50 values were also calculated. Best binding scores of -inhibitors against used enzymes were calculated as -12.095 kcal/mol, -12.775 kcal/mol, and -9.336 kcal/mol, respectively. While 5-oxo-triazole piperidine-4-carboxamide moieties have a critical role in the inhibition of AChE and GST enzymes, hydroxy benzyl moiety is important for BChE enzyme inhibition.


Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Heterocyclic Compounds/pharmacology , Piperidines/pharmacology , Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Glutathione Transferase/metabolism , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Piperidines/chemical synthesis , Piperidines/chemistry , Schiff Bases/chemical synthesis , Schiff Bases/chemistry , Schiff Bases/pharmacology , Spectrophotometry, Infrared
8.
Inorg Chem ; 60(17): 13051-13061, 2021 Sep 06.
Article En | MEDLINE | ID: mdl-34369147

The facile modification of the ligands in organometallic Ru(II)-arene complexes offers more opportunities to optimize their pharmacological profiles. Herein, three Ru(II)-arene complexes containing a glutathione S-transferase (GST) inhibitor (NBDHEX) in chelate ligand have been designed and synthesized in this study. In vitro results indicated that the ligation with NBDHEX significantly increased the activities and selectivities of the organometallic Ru(II)-arene complexes against tumor cells, especially complex 3, which was the most active compound among the tested compounds. DFT calculations and hydrolysis results demonstrated that complex 3 with more alkyl groups in the arene ligand has increased electron density at the Ru(II) center as compared with complexes 1 and 2, thus resulting in the improved hydrolysis rate, which may be responsible for its higher anticancer activity. Further studies showed that complexes 1-3 can cause the loss of the mitochondrial membrane potential and upregulate the expression of Bcl-2 and Bax in A549 cells, suggesting that complexes 1-3-induced cell death may be mediated via the mitochondrial apoptotic pathway. Thus, these findings suggested that simultaneous modification of the chelate ligands and arene rings in the organometallic Ru(II)-arene complexes is an effective way to improve their pharmacological properties.


Antineoplastic Agents/pharmacology , Coordination Complexes/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Coordination Complexes/chemical synthesis , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Molecular Structure , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Ruthenium/chemistry , S Phase Cell Cycle Checkpoints/drug effects , Up-Regulation/drug effects , bcl-2-Associated X Protein/metabolism
9.
Neurotoxicology ; 86: 85-93, 2021 09.
Article En | MEDLINE | ID: mdl-34314733

Parkinson's disease is characterized by dopamine dyshomeostasis and oxidative stress. The aldehyde metabolite of dopamine, 3,4-dihydroxyphenylacetaldehyde (DOPAL), has been reported to be cytotoxic and capable of protein modification. Protein modification by DOPAL has been implicated in the pathogenesis of Parkinson's disease, but the complete pathology is unknown. Our findings show that DOPAL modifies glutathione S-transferase (GST), an important enzyme in the antioxidant defense system. DOPAL, dopamine, and the metabolite 3,4-dihydroxyphenylacetic acid (DOPAC), inhibited the activity of GST isolated from N27 dopaminergic cells at an IC50 of 31.46 µM, 82.32 µM, and 260.0 µM, respectively. DOPAL, dopamine, and DOPAC inhibited commercially available equine liver GST at an IC50 of 23.72 µM, 32.17 µM, and 73.70 µM, respectively. This inhibition was time dependent and irreversible. 1 mM ʟ-cysteine or glutathione fully protected GST activity from DOPAL, DA, and DOPAC inhibition. 1 mM carnosine partially protected GST activity from DA inhibition. Furthermore, ʟ-cysteine was found to protect GST by forming a putative thiazolidine conjugate with DOPAL. We conclude that GST inactivation may be a part of the broader etiopathology of Parkinson's disease.


3,4-Dihydroxyphenylacetic Acid/analogs & derivatives , 3,4-Dihydroxyphenylacetic Acid/pharmacology , Dopamine/pharmacology , Dopaminergic Neurons/drug effects , Glutathione Transferase/antagonists & inhibitors , Animals , Cell Line , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dose-Response Relationship, Drug , Glutathione Transferase/metabolism , Horses , Rats
10.
Exp Parasitol ; 226-227: 108120, 2021.
Article En | MEDLINE | ID: mdl-34089701

The bio-efficacy of crude ethyl acetate extract, fractions and a compound phenyl acetic acid from the ethyl acetate extract of Streptomyces collinus was evaluated on Culex quinquefasciatus Say and Aedes aegypti L. mosquitoes (Diptera: Culicidae). The larvae were exposed to concentrations of 2.5, 5.0, 7.5 and 10.0 ppm for fractions and 0.5, 1.0, 1.5 and 2.0 ppm for compound. After 24 h, the larval mortality was assessed and the LC50 and LC90 values were calculated. Similarly, per cent ovicidal activity was calculated for eggs after 120 h post treatment for phenyl acetic acid. Among the eleven fractions screened, fraction 7 from the ethyl acetate extract of Streptomyces collinus exhibited good larvicidal activity against both mosquito species. The LC50 and LC90 values of fraction 7 were 4.42, 6.23 ppm against Cx. quinquefasciatus larvae and 5.13, 14.51 ppm against Ae. aegypti larvae, respectively. Further, the isolated compound, phenyl acetic acid from fraction 7 recorded 100% larvicidal activity at 2 ppm concentration with LC50 and LC90 values of 2.07, 4.87 ppm on Cx. quinquefasciatus larvae and 3.81, 9.87 ppm on Ae. aegypti larvae, respectively. Phenyl acetic acid presented 50.3% and 42.0% ovicidal activity against Cx. quinquefasciatus and Ae. aegypti eggs at 2 ppm concentration after 120 h post treatment. The compound, phenyl acetic acid could be used in mosquito control programme.


Aedes , Culex , Phenylacetates , Streptomyces/chemistry , Aedes/drug effects , Aedes/enzymology , Aedes/growth & development , Analysis of Variance , Animals , Biological Assay , Culex/drug effects , Culex/enzymology , Culex/growth & development , Esterases/antagonists & inhibitors , Glutathione Transferase/antagonists & inhibitors , India , Larva/drug effects , Lethal Dose 50 , Ovum/drug effects , Phenylacetates/chemistry , Phenylacetates/isolation & purification , Phenylacetates/pharmacology
11.
PLoS One ; 16(4): e0250704, 2021.
Article En | MEDLINE | ID: mdl-33909675

In this report, we extend our previous characterization of Dictyostelium discoideum glutathione S-transferase (DdGST) enzymes that are expressed in the eukaryotic model organism. Transcript profiling of gstA1-gstA5 (alpha class) genes in vegetative, log phase cells identified gstA2 and gstA3 with highest expression (6-7.5-fold, respectively) when compared to other gstA transcripts. Marked reductions in all gstA transcripts occurred under starvation conditions, with gstA2 and gstA3 exhibiting the largest decreases (-96% and -86.6%, respectively). When compared to their pre-starvation levels, there was also a 60 percent reduction in total GST activity. Glutathione (GSH) pull-down assay and mass spectroscopy detected three isozymes (DdGSTA1, DdGSTA2 and DdGSTA3) that were predominantly expressed in vegetative cells. Biochemical and kinetic comparisons between rDdGSTA2 and rDdGSTA3 shows higher activity of rDdGSTA2 to the CDNB (1-chloro-2,4-dinitrobenzene) substrate. RNAi-mediated knockdown of endogenous DdGSTA2 caused a 60 percent reduction in proliferation, delayed development, and altered morphogenesis of fruiting bodies, whereas overexpression of rDdGSTA2 enzyme had no effect. These findings corroborate previous studies that implicate a role for phase II GST enzymes in cell proliferation, homeostasis, and development in eukaryotic cells.


Dictyostelium/enzymology , Glutathione Transferase/metabolism , Cell Proliferation , Dictyostelium/growth & development , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , RNA Interference , RNA, Small Interfering/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Analysis, Protein
12.
Molecules ; 26(8)2021 Apr 20.
Article En | MEDLINE | ID: mdl-33924269

Human glutathione transferase A1-1 (hGSTA1-1) contributes to developing resistance to anticancer drugs and, therefore, is promising in terms of drug-design targets for coping with this phenomenon. In the present study, the interaction of anthraquinone and diazo dichlorotriazine dyes (DCTD) with hGSTA1-1 was investigated. The anthraquinone dye Procion blue MX-R (PBMX-R) appeared to interact with higher affinity and was selected for further study. The enzyme was specifically and irreversibly inactivated by PBMX-R, following a biphasic pseudo-first-order saturation kinetics, with approximately 1 mol of inhibitor per mol of the dimeric enzyme being incorporated. Molecular modeling and protein chemistry data suggested that the modified residue is the Cys112, which is located at the entrance of the solvent channel at the subunits interface. The results suggest that negative cooperativity exists upon PBMX-R binding, indicating a structural communication between the two subunits. Kinetic inhibition analysis showed that the dye is a competitive inhibitor towards glutathione (GSH) and mixed-type inhibitor towards 1-chloro-2,4-dinitrobenzene (CDNB). The present study results suggest that PBMX-R is a useful probe suitable for assessing by kinetic means the drugability of the enzyme in future drug-design efforts.


Anticarcinogenic Agents/chemistry , Coloring Agents/chemistry , Glutathione Transferase/genetics , Neoplasms/drug therapy , Triazines/chemistry , Amino Acid Sequence/genetics , Anticarcinogenic Agents/therapeutic use , Binding Sites/drug effects , Dinitrochlorobenzene/chemistry , Glutathione/antagonists & inhibitors , Glutathione/genetics , Glutathione Transferase/antagonists & inhibitors , Humans , Kinetics , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/drug effects
13.
Biometals ; 34(2): 393-406, 2021 04.
Article En | MEDLINE | ID: mdl-33528765

A series of Fe(II), Ni(II), and Pd(II) complexes were prepared with a novel Schiff base ligand containing pyridine moiety. The prepared compounds were characterized using FT-IR, 1H and 13 C NMR, UV-Vis, powder XRD, thermogravimetric analysis, mass spectra, magnetic susceptibility, and elemental analysis. The coordination geometry of Fe(II) and Ni(II) complexes were octahedral, where Fe(II) and Ni(II) metal ions were coordinated by an oxygen atom of the carbonyl group, a nitrogen atom of the azomethine moiety, and a phenolic oxygen atom. The Pd(II) complex had square planar geometry. All of the synthesized compounds were tested for their biochemical properties, including enzyme inhibition and antioxidant activities. According to the in vitro DPPH and FRAP antioxidant methods, the Schiff base ligand and its Fe(II)/Pd(II) complexes showed close antioxidant activities against the standards (BHA, BHT, ascorbic acid, and α-tocopherol). Enzyme inhibitions of the metal complexes were investigated against glutathione S-transferase (GST), acetylcholinesterase (AChE), and butyrylcholinesterase (BChE) enzymes. The best inhibition value (Ki) was observed for the Ni(II) complex against GST (2.63 ± 0.04 µM). Also, the Pd(II) complex showed the best inhibition value (10.17 ± 1.88 µM) against AChE. Molecular docking specified significant interactions at the active pockets of respective target enzymes. The Ni(II) complex exhibited good binding affinity against both BChE (- 9.0 kcal/mol and 9.36 ± 2.03 µM) and GST (- 7.0 kcal/mol and 2.63 ± 0.04 µM) enzymes.


Antioxidants/pharmacology , Coordination Complexes/pharmacology , Enzyme Inhibitors/pharmacology , Metals, Heavy/pharmacology , Molecular Docking Simulation , Pyridines/pharmacology , Acetylcholinesterase/metabolism , Antioxidants/chemical synthesis , Antioxidants/chemistry , Biphenyl Compounds/antagonists & inhibitors , Butyrylcholinesterase/metabolism , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fluorescence Recovery After Photobleaching , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/metabolism , Ligands , Metals, Heavy/chemistry , Molecular Structure , Picrates/antagonists & inhibitors , Pyridines/chemistry , Schiff Bases/chemistry , Schiff Bases/pharmacology
14.
J Med Chem ; 64(3): 1701-1712, 2021 02 11.
Article En | MEDLINE | ID: mdl-33529017

Glutathione transferase (GST P1-1) is a potential target for anticancer drugs. In this work, a series of 6-(7-nitro-2,1,3-benzoxadiazol-4-ylthio)hexanol (NBDHEX) derivatives as GST P1-1 inhibitors were designed, synthesized, and evaluated for their biological activity. Among the target compounds, 4n showed more selective inhibition toward GST P1-1 and GST M2-2, better water solubility, and more potent anticancer activities toward all the tested cancer cells (except for HOS) than its parent molecule. Detailed biological studies on the effect of 4n toward 143b cells revealed that 4n could arrest the cell cycle at the G2 phase and induced cell apoptosis in a dose-dependent manner. Like NBDHEX, 4n displayed good pharmacokinetic characteristics. An in vivo study on 143b xenograft models demonstrated that 4n could significantly reduce tumor growth in a dose-dependent manner, showing stronger antitumor activity than NBDHEX. Thus, 4n deserves to be further investigated as a potential antitumor agent for cancer therapy.


Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Oxadiazoles/chemistry , Animals , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Discovery , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/toxicity , G2 Phase/drug effects , Humans , Molecular Docking Simulation , Rats , Rats, Sprague-Dawley , Solubility , Xenograft Model Antitumor Assays
15.
Bioorg Chem ; 107: 104606, 2021 02.
Article En | MEDLINE | ID: mdl-33476865

In this study, new 1,2,3-triazole derivatives containing chalcone core (1-7) were synthesized. Obtained compounds were characterized by IR, 1H NMR, 13C NMR, and mass studies. Characterized compounds (1-7) inhibitory effects were tested against the glutathione S-transferase (GST), acetylcholinesterase (AChE), and Butyrylcholinesterase (BChE). Their Ki values were in the range of 5.88-11.13 µM on AChE, 5.08-15.12 µM on BChE, and 9.82-13.22 µM on GST. Remarkable inhibitory effects were obtained against three tested metabolic enzymes. Also, binding scores of the best-inhibitors against AChE, BChE, and GST enzymes were detected as -9.969 kcal/mol, -10.672 kcal/mol, and -8.832 kcal/mol, respectively. Isoindoline-1,3-dione and benzothiophene moieties played a critical role in the inhibition of AChE and BChE enzymes, respectively. Phenylene and triazole moieties had the most important interactions for inhibition of the GST enzyme. Therefore, in vivo and in silico results indicated that these compounds can be considered in drug design processes for the treatment of some diseases including Alzheimer's disease (AD), leukemia, and some type of cancer.


Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Glutathione Transferase/metabolism , Triazoles/chemistry , Acetylcholinesterase/chemistry , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Binding Sites , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/metabolism , Cholinesterase Inhibitors/therapeutic use , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Glutathione Transferase/antagonists & inhibitors , Humans , Molecular Docking Simulation , Protein Binding , Protein Structure, Tertiary , Structure-Activity Relationship , Triazoles/metabolism , Triazoles/therapeutic use
16.
Curr Drug Metab ; 22(4): 308-314, 2021.
Article En | MEDLINE | ID: mdl-33461457

BACKGROUND: The glutathione S-transferases (GSTs) are family of enzymes that are notable for their role in phase II detoxification reactions. Antibiotics have been reported to have several adverse effects on the activity of the enzymes in mammals. AIM: The aim of this study was the structural and biochemical characterization of rat erythrocyte GST and understanding the effects of gentamicin, clindamycin, cefazolin, ampicillin and scopolamine butylbromide on the activity of human erythrocyte GST using rat as a model. METHODS: The enzyme was purified by GSH-agarose affinity chromatography. In vitro GST enzyme activity was measured at 25°C using CDNB as a model substrate. IC50 of drugs was measured by activity % vs compound concentration graphs. Lineweaver Burk graphs were drawn to determine the inhibition type and Ki constants for the drugs. The structure of the enzyme was predicted via Protein Homology/analogy Recognition Engine. RESULTS: In this study, GST was purified from rat erythrocyte with a specific activity of 6.3 EU/mg protein, 44 % yield and 115 fold. Gentamicin and clindamycin inhibited the enzymatic activity with IC50 of 1.69 and 6.9 mM and Ki of 1.70 and 2.36 mM, respectively. Ampicillin and scopolamine butylbromide were activators of the enzyme, while the activity of the enzyme was insensitive to cefazolin. The enzyme was further characterized by homology modeling and sequence alignment revealing similarities with human GST. CONCLUSION: Collectively, it could be concluded that gentamicin and clindamycin are the inhibitors of erythrocyte GST.


Anti-Bacterial Agents/pharmacology , Glutathione Transferase/antagonists & inhibitors , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Enzyme Assays , Erythrocytes/enzymology , Glutathione Transferase/genetics , Glutathione Transferase/isolation & purification , Glutathione Transferase/metabolism , Humans , Inhibitory Concentration 50 , Male , Rats , Sequence Alignment , Sequence Homology, Amino Acid
17.
Bioorg Chem ; 107: 104524, 2021 02.
Article En | MEDLINE | ID: mdl-33317836

The synthesized Schiff Bases were reacted with formaldehyde and secondary amine such as 2,6-dimethylmorpholine to afford N-Mannich bases through the Mannich reaction. 3-Substitued-4-(4-hydroxybenzylidenamino)-4,5-dihydro-1H-1,2,4-triazol-5-ones (4) were treated with 2,6-dimethylmorpholine in the presence of formaldehyde to synthesize eight new 1-(2,6-dimethylmorpholino-4-yl-methyl)-3-substitued-4-(4-hydroxybenzylidenamino)-4,5-dihydro-1H-1,2,4-triazol-5-ones (4a-h). The structures of the synthesized eight new compounds were characterized using IR, 1H NMR, 13C NMR, and HR-MS spectroscopic methods. Synthesized compounds inhibitory activity determined against the acetylcholinesterase (AChE), butyrylcholinesterase (BChE), and glutathione S-transferase (GST) enzymes with Ki values in the range 25.23-42.19 µM for AChE, 19.37-34.22 µM for BChE, and 21.84-41.14 µM for GST, respectively. Binding scores of most active inhibitors against AChE, BChE, and GST enzymes were detected as -10.294 kcal/mol, -9.562 kcal/mol, and -7.112 kcal/mol, respectively. The hydroxybenzylidene moiety of the most active inhibitors caused to inhibition of the enzymes through hydrophobic interaction and hydrogen bond.


Cholinesterase Inhibitors/pharmacology , Mannich Bases/pharmacology , Morpholines/pharmacology , Schiff Bases/pharmacology , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Animals , Butyrylcholinesterase/chemistry , Butyrylcholinesterase/metabolism , Caco-2 Cells , Catalytic Domain , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/metabolism , Dogs , Drug Design , Enzyme Assays , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Madin Darby Canine Kidney Cells , Mannich Bases/chemical synthesis , Mannich Bases/metabolism , Molecular Docking Simulation , Morpholines/chemical synthesis , Morpholines/metabolism , Protein Binding , Schiff Bases/chemical synthesis , Schiff Bases/metabolism
18.
Anticancer Agents Med Chem ; 21(10): 1292-1300, 2021.
Article En | MEDLINE | ID: mdl-32951581

BACKGROUND: Indole-based heterocyclic compounds play important roles in pharmaceutical chemistry due to their unexpected biological and pharmacological properties. OBJECTIVE: Herein, we describe novel biological properties (antioxidant, antimicrobial and anti-cancer) of 3- bromo-1-ethyl-1H-indole (BEI) structure. METHODS: BEI was synthesized from 1-Methyl-2-phenylindole and N-bromosuccinimide and was characterized by using 1H and 13C NMR. Cytotoxicity was determined by MTT assay. Apoptosis analysis of BEI was determined by Arthur™ image-based Cytometer. Different methods were applied to assess the antioxidant activity of BEI. Molecular docking studies were conducted to determine the interactions of bonding between GST isozymes and BEI. RESULTS: According to the antioxidant and antimicrobial activity assays, BEI compound showed reduced total antioxidant activity compared to the Trolox standard, whereas it showed moderate antimicrobial activity against Aspergillus niger and Phytophora eryhtrospora. Notably, the BEI compound demonstrated substantial selective cytotoxicity for the first time towards cancer cell lines, and there existed a significant decrease in the percentage of live cells treated with BEI, in comparison to the control ones. Interestingly, BEI exhibited a promising glutathione S-transferase isozymes inhibition. CONCLUSION: The results of this study suggest that BEI seems to be a promising molecule to be used in the design of new anti-cancer agents that provide superiority to present commercial anti-cancer drugs.


Anti-Bacterial Agents/pharmacology , Antifungal Agents/pharmacology , Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Antifungal Agents/chemical synthesis , Antifungal Agents/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antioxidants/chemical synthesis , Antioxidants/chemistry , Apoptosis/drug effects , Bacteria/drug effects , Benzothiazoles/antagonists & inhibitors , Biphenyl Compounds/antagonists & inhibitors , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Fungi/drug effects , Glutathione Transferase/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Microbial Sensitivity Tests , Molecular Structure , Picrates/antagonists & inhibitors , Structure-Activity Relationship , Sulfonic Acids/antagonists & inhibitors
19.
Med Chem ; 17(4): 332-343, 2021.
Article En | MEDLINE | ID: mdl-32242785

BACKGROUND: The 26kDa glutathione transferase (GST, EC 2.5.1.18) from Schistosoma japonicum (SjGST) is recognized as the major detoxification enzyme of S. japonicum, a pathogenic helminth causing schistosomiasis. OBJECTIVE: In the present study, the interaction of the chlorotriazine dye Cibacron blue 3GA (CB3GA) and its structural analogues with SjGST was investigated. The work aimed to shed light on the non-substrate ligand-binding properties of the enzyme. METHODS: Kinetic inhibition analysis, affinity labelling experiments and molecular modelling studies were employed. RESULTS: The results showed that CB3GA is a potent inhibitor (IC50 0.057 ± 0.003 µM) towards SjGST. The enzyme was specifically and irreversibly inactivated by the dichlorotriazine-analogue of CB3GA (IC50 0.190 ± 0.024 µM), following a biphasic pseudo-first-order saturation kinetics with approximately 1 mol of inhibitor per mol of the dimeric enzyme being incorporated. All other monochlorotriazine analogues behave as reversible inhibitors with lower inhibition potency (IC50 5.2-82.3 µM). Kinetic inhibition studies, together with molecular modelling and molecular dynamics simulations, established that the CB3GA binding site overlaps both the G- and H-sites. Both hydrophobic/ polar interactions, as well as steric effects, have decisive roles in determining the inhibitory strength of CB3GA and its analogues. CONCLUSION: The results of the present study might be useful in future drug design and development efforts towards SjGST.


Glutathione Transferase/metabolism , Helminth Proteins/metabolism , Schistosoma japonicum/enzymology , Triazines/metabolism , Animals , Enzyme Assays , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/chemistry , Helminth Proteins/antagonists & inhibitors , Helminth Proteins/chemistry , Kinetics , Ligands , Molecular Docking Simulation , Protein Binding , Triazines/chemistry
20.
J Vis Exp ; (164)2020 10 10.
Article En | MEDLINE | ID: mdl-33104076

Glutathione S-transferases (GSTs) are metabolic enzymes responsible for the elimination of endogenous or exogenous electrophilic compounds by glutathione (GSH) conjugation. In addition, GSTs are regulators of mitogen-activated protein kinases (MAPKs) involved in apoptotic pathways. Overexpression of GSTs is correlated with decreased therapeutic efficacy among patients undergoing chemotherapy with electrophilic alkylating agents. Using GST inhibitors may be a potential solution to reverse this tendency and augment treatment potency. Achieving this goal requires the discovery of such compounds, with an accurate, quick, and easy enzyme assay. A spectrophotometric protocol using 1-chloro-2,4-dinitrobenzene (CDNB) as the substrate is the most employed method in the literature. However, already described GST inhibition experiments do not provide a protocol detailing each stage of an optimal inhibition assay, such as the measurement of the Michaelis-Menten constant (Km) for CDNB or indication of the employed enzyme concentration, crucial parameters to assess the inhibition potency of a tested compound. Hence, with this protocol, we describe each step of an optimized spectrophotometric GST enzyme assay, to screen libraries of potential inhibitors. We explain the calculation of both the half-maximal inhibitory concentration (IC50) and the constant of inhibition (Ki)-two characteristics used to measure the potency of an enzyme inhibitor. The method described can be implemented using a pool of GSTs extracted from cells or pure recombinant human GSTs, namely GST alpha 1 (GSTA1), GST mu 1 (GSTM1) or GST pi 1 (GSTP1). However, this protocol cannot be applied to GST theta 1 (GSTT1), as CDNB is not a substrate for this isoform. This method was used to test the inhibition potency of curcumin using GSTs from equine liver. Curcumin is a molecule exhibiting anti-cancer properties and showed affinity towards GST isoforms after in silico docking predictions. We demonstrated that curcumin is a potent competitive GST inhibitor, with an IC50 of 31.6 ± 3.6 µM and a Ki of 23.2 ± 3.2 µM. Curcumin has potential to be combined with electrophilic chemotherapy medication to improve its efficacy.


Cytosol/enzymology , Drug Evaluation, Preclinical , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Glutathione Transferase/antagonists & inhibitors , Spectrophotometry/methods , Animals , Curcumin/pharmacology , Dinitrobenzenes/metabolism , Ethacrynic Acid/pharmacology , Glutathione/metabolism , Glutathione Transferase/metabolism , Horses , Inhibitory Concentration 50 , Isoenzymes/metabolism , Kinetics , Liver/enzymology , Substrate Specificity/drug effects
...