Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Comput Biol Chem ; 110: 108052, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38492557

ABSTRACT

Alpha-glucosidase (maltase, sucrase, isomaltase and glucoamylase) activities which are involved in carbohydrate metabolism are present in human intestinal maltase-glucoamylase (MGAM) and sucrase-isomaltase (SI). Hence, these proteins are important targets to identify drugs against postprandial hyperglycemia thereby for diabetes. To find natural-based drugs against MGAM and SI, Artocarpus heterophyllus leaf was explored for MGAM and SI inhibition in in vitro and in silico. A. heterophyllus leaf aqueous active fraction (AHL-AAF) was prepared using Soxhlet extraction followed by silica column chromatography. The phytoconstituents of AHL-AAF were determined using LC-ESI-MS/MS. AHL-AAF showed dose-dependent and mixed inhibition against maltase (IC50 = 460 µg/ml; Ki = 300 µg/ml), glucoamylase (IC50 = 780 µg/ml; Ki = 480 µg/ml), sucrase (IC50 = 900 µg/ml, Ki = 504 µg/ml) and isomaltase (IC50 = 860 µg/ml, Ki = 400 µg/ml). AHL-AAF phytoconstituents interaction with N-terminal (Nt) and C-terminal (Ct) subunits of human MGAM and SI was analyzed using induced-fit docking, molecular dynamics (MD), and binding free energy calculation. In docking studies, rhamnosyl hexosyl methyl quercetin (RHMQ), P-coumaryl-O-16-hydroxy palmitic acid (PCHP), and spirostanol interacted with active site amino acids of human MGAM and SI. Among these RHMQ stably interacted with all the subunits (Nt-MGAM, Ct-MGAM, Nt-SI and Ct-SI) whereas PCHP with Ct-MGAM and Nt-SI during MD analysis. In molecular docking, the docking score of RHMQ with NtMGAM, CtMGAM, NtSI and CtSI was -8.48, -12.88, -11.98 and -11.37 kcal/mol. The docking score of PCHP for CtMGAM and NtSI was -8.59 and -8.4 kcal/mol, respectively. After MD simulation, the root mean square deviation (RMSD) and root mean square fluctuation (RMSF) values further confirmed the stable protein-ligand interaction. The RMSD value of all the complexes were around 2.5 Šand the corresponding RMSF values were also quite low. In MM/GBSA analysis, the involvement of Van der Waals and lipophilic energy in the protein/ligand interactions are understood. Further binding free energy for Nt-MGAM-PCHP, Nt-MGAM-RHMQ, Nt-SI-PCHP, Nt-SI-RHMQ, Ct-MGAM-PCHP, Ct-MGAM-RHMQ and Ct-SI-RHMQ complexes was found to be -24.94, -46.60, -46.56, -44.48, -40.3, -41.86 and -19.39 kcal/mol, respectively. Altogether, AHL-AAF showed inhibition of α-glucosidase activities of MGAM and SI. AHL-AAF could be further studied for its effect on diabetes in in vivo.


Subject(s)
Artocarpus , Molecular Docking Simulation , Artocarpus/chemistry , Humans , alpha-Glucosidases/metabolism , alpha-Glucosidases/chemistry , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Molecular Dynamics Simulation , Glucan 1,4-alpha-Glucosidase/metabolism , Glucan 1,4-alpha-Glucosidase/antagonists & inhibitors , Glucan 1,4-alpha-Glucosidase/chemistry , Plant Leaves/chemistry , Sucrase-Isomaltase Complex/antagonists & inhibitors , Sucrase-Isomaltase Complex/metabolism , Sucrase-Isomaltase Complex/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Structure-Activity Relationship , Animals , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology
2.
Comput Biol Chem ; 108: 107996, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38061170

ABSTRACT

Targeting multiple factors such as oxidative stress, alpha glucosidase and acetylcholinesterase (AChE) are considered advantageous for the treatment of diabetes and diabetes associated-cognitive dysfunction. In the present study, Hibiscus rosa-sinensis flowers anthocyanin-rich extract (HRA) was prepared. Phytochemical analysis of HRA using LC-ESI/MS/MS revealed the presence of various phenolic acids, flavonoids and anthocyanins. HRA showed in vitro antioxidant activity at low concentrations. HRA inhibited all the activities of mammalian glucosidases and AChE activity. The IC50 value of HRA for the inhibition of maltase, sucrase, isomaltase, glucoamylase and AChE was found to be 308.02 ± 34.25 µg/ml, 287.8 ± 19.49 µg/ml, 424.58 ± 34.75 µg/ml, 408.94 ± 64.82 µg/ml and 264.13 ± 30.84 µg/ml, respectively. Kinetic analysis revealed mixed-type inhibition against all the activities except for glucoamylase (competitive) activity. In silico analysis confirmed the interaction of two active constituents cyanidin 3-sophoroside (CS) and quercetin 3-O-sophoroside (QS) with four subunits, n-terminal and c-terminal subunits of human maltase-glucoamylase and sucrase-isomaltase as well as with AChE. Molecular dynamics simulation, binding free energy calculation, DCCM, PCA, PCA-based free energy surface analysis ascertained the stable binding of CS and QS with target proteins studied. HRA could be used as complementary therapy for diabetes and cognitive improvement.


Subject(s)
Flowers , Glucosidases , Hibiscus , Animals , Humans , Acetylcholinesterase/metabolism , alpha-Glucosidases/metabolism , Anthocyanins/pharmacology , Diabetes Mellitus , Flowers/chemistry , Glucan 1,4-alpha-Glucosidase/antagonists & inhibitors , Glucan 1,4-alpha-Glucosidase/metabolism , Glucosidases/antagonists & inhibitors , Hibiscus/chemistry , Kinetics , Oligo-1,6-Glucosidase/antagonists & inhibitors , Plant Extracts/pharmacology , Sucrase/antagonists & inhibitors , Tandem Mass Spectrometry , Glycoside Hydrolase Inhibitors/pharmacology , Phytochemicals/pharmacology
3.
Mol Divers ; 27(6): 2741-2766, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36547813

ABSTRACT

Severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) pathogenesis is initiated by the binding of SARS-CoV-2 spike (S) protein with the angiotensin-converting enzyme 2 receptor (ACE2R) on the host cell surface. The receptor-binding domain (RBD) of the S protein mediates the binding and is more prone to mutations resulting in the generation of different variants. Recently, molecules with the potential to inhibit the interaction of S protein with ACE2R have been of interest due to their therapeutic value. In this context, the present work was performed to identify potential RBD binders from the Indian medicinal plant's phytochemical database through virtual screening, molecular docking, and molecular dynamic simulation. Briefly, 1578 compounds filtered from 9596 phytochemicals were chosen for screening against the RBD of the native SARS-CoV-2 S protein. Based on the binding energy, the top 30 compounds were selected and re-docked individually against the native and five variants of concern (VOCs: alpha, beta, gamma, delta, and omicron) of SARS-CoV-2. Four phytochemicals, namely withanolide F, serotobenine, orobanchol, and gibberellin A51, were found to be potential RBD binders in native and all SARS-CoV-2 VOCs. Among the four, withanolide F exhibited lower binding energy (- 10.84 to - 8.56 kcal/mol) and better ligand efficiency (- 0.3 to - 0.25) against all forms of RBD and hence was subjected to a 100 ns MD simulation which confirmed its stringent binding to the RBDs in native and VOCs. The study prioritizes withanolide F as a prospective COVID-19 (Coronavirus disease) therapeutic agent based on the observations. It warrants deeper investigations into the four promising leads for understanding their precise therapeutic value.


Subject(s)
COVID-19 , Withanolides , Humans , SARS-CoV-2 , Molecular Docking Simulation , Protein Binding , Molecular Dynamics Simulation
4.
Appl Biochem Biotechnol ; 194(12): 5827-5847, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35819687

ABSTRACT

In recent years, derivatives of natural compounds are synthesized to increase the bioavailability, pharmacology, and pharmacokinetics properties. The naphthoquinone, plumbagin (PLU), is well known for its anticancer activity. However, the clinical use of PLU is hindered due to its toxicity. Previous reports have shown that modification of PLU at 5'-hydroxyl group has reduced its toxicity towards normal cell line. In accordance, in the present study, 5'-hydroxyl group of PLU was esterified with S-allyl cysteine (SAC) to obtain PLU-SAC ester. The drug-likeness of PLU-SAC was understood by in silico ADME analysis. PLU-SAC was characterized by UV-visible spectroscopy, mass spectroscopy, and nuclear magnetic resonance (NMR) spectroscopy. Molecular docking and dynamics simulation analysis revealed the interaction of PLU-SAC with proteins of interest in cancer therapy such as human estrogen receptor α, tumor protein p53 negative regulator mouse double minute 2, and cyclin-dependent kinase 2. MMGBSA calculation showed the favorable binding energy which in turn demonstrated the stable binding of PLU-SAC with these proteins. PLU-SAC showed apoptosis in breast cancer cell line (MCF-7) by inducing oxidative stress, disturbing mitochondrial function, arresting cells at G1 phase of cell cycle, and initiating DNA fragmentation. However, PLU-SAC did not show toxicity towards normal Vero cell line. PLU-SAC was synthesized and structurally characterized, and its anticancer activity was determined by in silico and in vitro analysis.


Subject(s)
Esters , Naphthoquinones , Humans , Mice , Animals , Molecular Docking Simulation , Esters/pharmacology , Naphthoquinones/pharmacology , Naphthoquinones/chemistry , Cysteine/chemistry , Apoptosis , Cell Line, Tumor
5.
Arch Physiol Biochem ; : 1-9, 2022 Apr 28.
Article in English | MEDLINE | ID: mdl-35482540

ABSTRACT

Purpose: High glucose (HG)-induced oxidative stress is associated with apoptosis in pancreatic ß-cells. The protective effect of astaxanthin-s-allyl cysteine diester (AST-SAC) against HG-induced oxidative stress in pancreatic ß-cells (ßTC-tet cell line) in in vitro was studied.Materials and Methods: ßTC-tet cell line was exposed to HG in the presence and absence of AST-SAC. Various parameters such as cell viability, reactive oxygen species generation, mitochondrial membrane potential, DNA fragmentation and expression of proteins involved in apoptosis [p53, B-cell lymphoma 2 (Bcl-2), Bcl-2 associated X (Bax), cytochrome c and caspase 3] were studied.Results: Pre-treatment of ßTC-tet cells with AST-SAC (4, 8 and 12 µg/ml) in the presence of HG (25 mM) protected the viability of the cells in a dose-dependent manner. AST-SAC treatment mitigated the oxidative stress thereby preventing the mitochondrial dysfunction, DNA damage and apoptosis in ßTC-tet cells against HG toxicity. Treatment with AST-SAC prevented the increased expression of p53 under HG conditions. Further, AST-SAC treatment maintained the level of pro-apoptotic (Bax, cleaved caspase-3 and cytochrome c) and anti-apoptotic (Bcl-2) proteins to that of the control level under HG exposed conditions in ßTC-tet cells.Conclusion: Altogether, AST-SAC alleviated HG-induced oxidative damage and apoptosis in pancreatic ß-cells by enhancing the antioxidant status and altering apoptotic-related protein expression.

6.
J Biomol Struct Dyn ; 40(22): 11511-11525, 2022.
Article in English | MEDLINE | ID: mdl-34344261

ABSTRACT

Nonivamide (NOV), less pungent analogue of capsaicin present in various Capsicum species is known for various biological properties. S-allyl cysteine (SAC) abundantly present in aged garlic extract is gaining importance for anticancer property. NOV was esterified with SAC to increase the biological activity. In silico ADME analysis revealed the drug-likeness of NOV-SAC. Molecular docking and dynamics simulation analysis were done to understand the interaction of NOV-SAC with therapeutic target proteins (human estrogen receptor α, tumo protein negative regulator mouse double minute 2, B-cell lymphoma 2 and cyclin-dependent kinase 2) to treat cancer. NOV-SAC interacted with these proteins stably with favorable binding energy which was calculated through MMGBSA method. In line with in silico results, NOV-SAC showed antiproliferative activity against breast cancer cell line (MCF-7). NOV-SAC treatment increased ROS generation, decreased the antioxidant level, arrested cells at G1/S phase, disrupted mitochondrial membrane potential and initiated DNA fragmentation. The expression of p53 is increased by NOV-SAC treatment, in concordance the ratio of Bcl-2/Bax was decreased. Altogether, NOV-SAC was synthesized for the first time and it induced apoptosis in MCF-7 cells through triggering ROS generation and increasing the expression of p53. The in silico results has been mirrored in in vitro analysis of NOV-SAC against cancer cell line.Communicated by Ramaswamy H. Sarma.


Subject(s)
Capsaicin , Tumor Suppressor Protein p53 , Mice , Humans , Animals , Aged , Reactive Oxygen Species/metabolism , Capsaicin/pharmacology , Molecular Docking Simulation , Antioxidants/pharmacology , Apoptosis , Cysteine/chemistry , MCF-7 Cells
7.
Neurotoxicology ; 86: 114-124, 2021 09.
Article in English | MEDLINE | ID: mdl-34339762

ABSTRACT

Neuroprotective effect of astaxanthin-s-allyl cysteine diester (AST-SAC) against high glucose (HG)-induced oxidative stress in in vitro and cognitive decline under diabetes conditions in in vivo has been explored. Pretreatment of AST-SAC (5, 10 and 15 µM) dose-dependently preserved the neuronal cells (SH-SY5Y) viability against HG toxicity through i) decreasing oxidative stress (decreasing reactive oxygen species generation and increasing endogenous antioxidants level); ii) protecting mitochondrial function [oxidative phosphorylation (OXPHOS) complexes activity and mitochondrial membrane potential (MMP)]; and iii) decreasing p53 level thereby subsequently decreasing the level of apoptotic marker proteins. Male Spraque-Dawley rats were orally administered AST-SAC (1 mg/kg/day) for 45 days in streptozotocin-induced diabetes mellitus (DM) rats. AST-SAC administration prevented the loss of spatial memory in DM rats as determined using the novel object location test. AST-SAC administration alleviated the DM-induced injury in brain such as increased cholinesterases activity, elevated oxidative stress and mitochondrial dysfunction. Altogether, the results from the present study demonstrated that AST-SAC averted the neuronal apoptosis and preserved the cognitive function against HG toxicity under DM conditions.


Subject(s)
Cognitive Dysfunction/metabolism , Cysteine/analogs & derivatives , Diabetes Mellitus, Experimental/metabolism , Mitochondria/metabolism , Oxidative Stress/drug effects , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/drug therapy , Cysteine/pharmacology , Cysteine/therapeutic use , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , Dose-Response Relationship, Drug , Glucose/toxicity , Humans , Male , Mitochondria/drug effects , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/metabolism , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
8.
Int J Biol Macromol ; 185: 750-760, 2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34216669

ABSTRACT

Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are considered important target for drug design against Alzheimer's disease. In the present study in silico analysis; theoretical analysis of biointerface between ligand and interacting amino acid residues of proteins; and in vitro analysis of enzyme inhibition kinetics were carried out to delineate the inhibitory property of amine compounds against AChE/BChE. High throughput virtual screening of amine compounds identified three compounds (2-aminoquinoline, 2-aminobenzimidazole and 2-amino-1-methylbenzimidazole) that best interacted with AChE/BChE. Molecular docking analysis revealed the interaction of these compounds in the active site gorge of AChE/BChE, in particular with amino acid residues present in the peripheral anionic site. Molecular dynamics simulation confirmed the stable binding of these compounds with AChE/BChE. Binding energy calculated through MMGBSA method identified the non-covalent interactions (electrostatic and Van der Waals interactions) have contributed to the stable binding of the amine compounds with the AChE/BChE. Biointerface between amine compounds and AChE/BChE were visualized through Hirshfeld surface analysis. The inter-fragment interaction energies for the possible contacts between amine compounds and amino acid residues were carried out for the first time. All the amine compounds showed mixed-type of inhibition with moderate Ki value in in vitro analysis.


Subject(s)
Acetylcholinesterase/chemistry , Amines/pharmacology , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/pharmacology , Acetylcholinesterase/metabolism , Amines/chemistry , Binding Sites , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/chemistry , Computer Simulation , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Kinetics , Models, Molecular , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Structure-Activity Relationship
9.
Int J Biol Macromol ; 160: 623-631, 2020 Oct 01.
Article in English | MEDLINE | ID: mdl-32473219

ABSTRACT

Inhibition of pancreatic lipase (PL) is considered one of the important therapeutic interventions against obesity. In the present study, the inhibition of porcine (mammalian) PL (PPL) by two tripeptides glutathione (GSH) and s-allyl glutathione (SAG) was studied. In vitro kinetic analysis was done to determine the inhibition of GSH and SAG against PPL. The binding of GSH and SAG with PPL was elucidated by fluorescence spectroscopy analysis. Docking and molecular dynamics (MD) simulation analysis was carried out to understand the intermolecular interaction between both GSH and SAG with PPL as well as human PL (HPL). Both GSH and SAG inhibited PPL in mixed non-competitive manner. The IC50 value for GSH and SAG against PPL was found to be 2.97 and 6.4 mM, respectively. Both GSH and SAG quenched the intrinsic fluorescence of PPL through static quenching that is through forming complex with the PPL. SAG and GSH interacted with amino acids involved in catalysis of both PPL and HPL. MD simulation showed interactions of SAG and GSH with both PPL and HPL were stable. These results would lead to the further studies and application of GSH and SAG against obesity through inhibition of PL.


Subject(s)
Glutathione/pharmacology , Lipase/antagonists & inhibitors , Pancreas/drug effects , Amino Acids/metabolism , Animals , Catalysis/drug effects , Humans , Kinetics , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Spectrometry, Fluorescence/methods , Swine
10.
Int J Biol Macromol ; 148: 696-703, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-31954795

ABSTRACT

The isolation and purification of active components from the brown algae Sargassum.wightii is highly limited. In the present study, fucoxanthin was purified from S. wightii using simple methods. Ethyl acetate fraction obtained by Soxhlet extraction contained high concentration of fucoxanthin. Fucoxanthin-rich fraction was further subjected to open silica column chromatography and thin layer chromatography to obtain purified fucoxanthin. Purified fucoxanthin showed in vitro antioxidant activity. Fucoxanthin showed inhibition of angiotensin I-converting enzyme (ACE) with half maximal inhibitory value of 822.64 ± 17.69 µM. Kinetic analysis revealed mixed non-competitive inhibition with inhibitory constant of 600 µM for fucoxanthin against ACE. Molecular docking analysis showed the interaction of fucoxanthin with amino acids and zinc ion present in the active site of the human ACE. Molecular dynamics analysis demonstrated the stability of the fucoxanthin and ACE complex in in silico. These results show that S. wightii may be used as food ingredient to overcome hypertension.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/chemistry , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Peptidyl-Dipeptidase A/metabolism , Sargassum/chemistry , Xanthophylls/chemistry , Xanthophylls/pharmacology , Animals , Antioxidants/chemistry , Antioxidants/pharmacology , Computer Simulation , Kinetics , Molecular Docking Simulation , Rabbits
11.
Int J Biol Macromol ; 140: 1147-1157, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-31442505

ABSTRACT

In Alzheimer's disease (AD) and diabetes-associated cognitive decline, the acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activity is increased. AChE exists as different globular molecular forms: tetramer (G4), dimer (G2) and monomer (G1). In adult brain, G4 form is abundant however in AD, the ratio of lower molecular forms (G1) to G4 form increased. Hence, the present study delineated the inhibition of novel astaxanthin-s-allyl cysteine (AST-SAC) against BChE and various molecular forms of AChE. Cobra venom, human erythrocyte and Electrophorus electricus was used as source of G1, G2 and G4 form of AChE. AST-SAC showed inhibition against G1 (IC50 = 0.72 µM, competitive, Ki = 0.66 µM), G2 (IC50 = 0.65 µM, mixed, Ki = 0.50 µM) and G4 (IC50 = 0.67 µM, competitive, Ki = 0.67 µM) form of AChE. AST-SAC inhibited human brain AChE (IC50 = 0.84 µM, competitive, Ki = 0.53 µM) and human serum BChE (IC50 = 0.80 µM, competitive, Ki = 0.58 µM). In silico analysis revealed the interaction of AST-SAC with the amino acids present in peripheral anionic and catalytic site of human AChE and BChE. Molecular dynamics simulation confirmed the stable interaction of AST-SAC in the active site gorge of AChE and BChE.


Subject(s)
Acetylcholinesterase/metabolism , Butyrylcholinesterase/metabolism , Cholinesterase Inhibitors/pharmacology , Cysteine/analogs & derivatives , Acetylcholinesterase/chemistry , Animals , Brain/enzymology , Butyrylcholinesterase/blood , Butyrylcholinesterase/chemistry , Cholinesterase Inhibitors/chemistry , Computer Simulation , Cysteine/chemistry , Cysteine/pharmacology , Humans , Molecular Dynamics Simulation , Xanthophylls/chemistry , Xanthophylls/pharmacology
12.
Artif Cells Nanomed Biotechnol ; 47(1): 56-63, 2019 Dec.
Article in English | MEDLINE | ID: mdl-30669860

ABSTRACT

s-allyl glutathione (SAG) an analogue of glutathione is explored for its antioxidative and liver protection property in recent years. Selenium nanoparticles (Sh-SeNPs) were synthesized using medicinal plant Spermacoce hispida and conjugated with SAG (SAG-Sh-SeNPs). SAG-Sh-SeNPs and Sh-SeNPs were characterized using by Fourier transform infrared spectroscopy, Transmission electron microscopy, Energy dispersive X-ray analysis, X-ray diffraction analysis and zeta potential analysis. SAG-Sh-SeNPs and Sh-SeNPs were evaluated against acetaminophen (APAP)-induced liver and kidney injury in rat. Pretreatment of NPs attenuated the APAP toxicity-induced elevation of kidney and liver injury markers in the blood circulation. Histological observation showed that NPs pretreatment protected the morphology of liver and kidney tissue. SAG-Sh-SeNPs showed enhanced protection against APAP toxicity in comparison to Sh-SeNPs due to synergistic effect of SAG and Sh-SeNPs. SAG-Sh-SeNPs protected the liver and kidney against APAP toxicity through reducing oxidative stress, enhancing endogenous antioxidants and protecting mitochondrial functions.


Subject(s)
Acetaminophen/toxicity , Glutathione/chemistry , Kidney/drug effects , Liver/drug effects , Nanoparticles/chemistry , Rubiaceae/chemistry , Selenium/pharmacology , Animals , Antioxidants/metabolism , Apoptosis/drug effects , Cytoprotection/drug effects , Green Chemistry Technology , Kidney/cytology , Kidney/metabolism , Liver/cytology , Liver/metabolism , Male , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Selenium/chemistry
13.
Biochimie ; 154: 1-9, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30059711

ABSTRACT

S-allyl cysteine (SAC) is known for its various beneficial effects such as neuroprotection and immunomodulation. The beneficial effect of SAC against gout has not been explored. The present study aims to describe the two roles of SAC: (1) inhibitory effect against xanthine oxidase (XO) enzyme activity; and (2) anti-inflammatory property against MSU crystal-induced gouty inflammation in rat. The inhibitory effect of SAC against bovine XO enzyme activity was determined in vitro. In silico analysis was carried out to determine the intermolecular interaction between SAC and bovine XO. MSU crystal was injected in the right paw of the rat to induce gouty inflammation. SAC (40 mg/kg body weight) and colchicine (positive control; 1 mg/kg body weight) was given for 3 days. At the end of the treatment, the oxidative stress, antioxidant parameters and mitochondrial function were determined in the ankle joint tissue. The concentration of inflammatory cytokines such as TNF-α and IL-1ß was measured in the serum using ELISA. SAC inhibited (IC50 value, 33 µg/ml) XO enzyme activity in a competitive mode with corresponding Ki value of 4 µg/ml. In silico analysis predicted the interaction of SAC with the amino acids such as Arg880, Phe798, Phe914 and Phe1009 of XO enzyme. The root mean square deviation, root mean square fluctuation and free energy calculation values confirmed the stable SAC-XO interaction. The inhibition of SAC on XO enzyme activity in in vivo was further confirmed by silkworm model. SAC through reducing oxidative stress, enhancing antioxidants, protecting mitochondrial function has shown anti-inflammatory effect against MSU crystal-induced gout which was observed as reduced level of inflammatory markers in the serum. The medicinal potential of SAC as a preventive agent through its XO inhibitory property as well as curative agent through its anti-inflammatory property against gout has been understood from the present study.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Cysteine/analogs & derivatives , Gout Suppressants , Molecular Docking Simulation , Xanthine Oxidase , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cattle , Cysteine/chemistry , Cysteine/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Gout Suppressants/chemistry , Gout Suppressants/pharmacology , Humans , Milk , Rats, Sprague-Dawley , Xanthine Oxidase/antagonists & inhibitors , Xanthine Oxidase/chemistry , Xanthine Oxidase/metabolism
14.
Int J Biol Macromol ; 118(Pt A): 252-262, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-29885400

ABSTRACT

In humans, alpha-glucosidase activity is present in sucrase-isomaltase (SI) and maltase-glucoamylase (MGAM). α-glucosidase is involved in the hydrolyses of disaccharide into monosaccharides and results in hyperglycemia. Subsequently chronic hyperglycemia induces oxidative stress and ultimately leads to the secondary complications of diabetes. Hence, identifying compounds with dual beneficial activity such as efficient antioxidant and α-glucosidase inhibition property has attracted the attention in recent years. Keeping these views, in the present study astaxanthin (AST; a natural antioxidant present in marine microalgae) was biconjugated with allyl sulfur amino acid such as s-allyl cysteine (SAC). The synthesized AST-SAC (with molecular weight of 883.28) was characterized using UV-visible spectrophotometer, ESI-MS, and NMR analysis. AST-SAC showed potent antioxidant property in vitro. AST-SAC inhibited Saccharomyces cerevisiae α-glucosidase (IC50 = 3.98 µM; Ki = 1 µM) and mammalian α-glucosidase [rat intestinal maltase (IC50 = 6.4 µM; Ki = 1.3 µM) and sucrase (IC50 = 1.6 µM; Ki = 0.18 µM)] enzyme activity in a dose-dependent manner. Kinetic analysis revealed that AST-SAC inhibited all the α-glucosidases in a competitive mode. In silico analysis determined the interaction of AST-SAC with the amino acids present in the active site of S. cerevisiae and human (MGAM and SI) α-glucosidases.


Subject(s)
Cysteine/analogs & derivatives , alpha-Glucosidases/chemistry , Animals , Antioxidants/chemistry , Catalytic Domain , Computer Simulation , Cysteine/chemistry , Humans , Kinetics , Microalgae/chemistry , Molecular Docking Simulation , Rats , Saccharomyces cerevisiae/enzymology , Xanthophylls/biosynthesis , Xanthophylls/chemistry
15.
Environ Toxicol Pharmacol ; 58: 21-28, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29278860

ABSTRACT

A simple analogue of well known natural antioxidant glutathione (GSH) called S-allyl-glutathione (SAG) was evaluated against carbon tetrachloride (CCl4)-induced oxidative stress liver injury in rat. Pretreatment of SAG attenuated the CCl4-toxicity induced elevation of liver injury markers such as enzymes (AST, ALT, GGT, ALP and LDH) and bilirubin in the blood circulation. Such protective effect of SAG resulted in preservation of liver function observed as normal level of total protein and albumin in plasma as well as inhibition of dyslipidemia in liver. In addition, in silico analysis has proved that SAG has strong affinity with the amino acids present in active site of the human cytochrome P450 2E1 and 3A4. Three important mechanisms provided by SAG such as scavenging of reactive oxidants, replenishing of endogenous antioxidants (SOD, catalase, GPx, GSH and vitamin C) and protection of mitochondrial function (oxidative phosphorylation complex activities) are involved in the optimal function of liver against CCl4-toxicity.


Subject(s)
Antioxidants/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Glutathione/analogs & derivatives , Glutathione/therapeutic use , Animals , Antioxidants/pharmacology , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/metabolism , Glutathione/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Rats, Sprague-Dawley
16.
Metab Brain Dis ; 33(1): 151-159, 2018 02.
Article in English | MEDLINE | ID: mdl-29086235

ABSTRACT

Increased accumulation of endogenous neurotoxin quinolinic acid has been found in various neurodegenerative diseases. Oxidative stress caused by quinolinic acid is considered as imperative factor for its toxicity. Asiatic acid, a natural triterpene is widely studied for its various medicinal values. In the present study the effects of asiatic acid in preventing the cognitive impairment and oxidative stress caused by quinolinic acid was investigated. Male Spraque-Dawley rats were orally administered asiatic acid (30 mg/kg/day) for 28 days, while quinolinic acid toxicity-induced animals received quinolinic acid (1.5 mmol/kg/day) from day 15 to day 28 for 14 days. Asiatic acid administration prevented the loss of spatial memory caused due to quinolinic acid-induced toxicity as determined using the novel object location test. In addition, asiatic acid administration alleviated the deleterious effect of quinolinic acid in brain such as increased oxidative stress, decreased antioxidant status and mitochondrial oxidative phosphorylation dysfunction. These data demonstrate that asiatic acid through its potent antioxidant and cognition enhancement property prevented the neuronal impairments caused by quinolinic acid.


Subject(s)
Cognition/drug effects , Cognitive Dysfunction/prevention & control , Oxidative Stress/drug effects , Pentacyclic Triterpenes/pharmacology , Spatial Memory/drug effects , Animals , Antioxidants/pharmacology , Cognitive Dysfunction/drug therapy , Lipid Peroxidation/drug effects , Male , Neurodegenerative Diseases/drug therapy , Quinolinic Acid/pharmacology , Rats, Sprague-Dawley
17.
J Biosci Bioeng ; 116(2): 152-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23474097

ABSTRACT

A bifunctional xylosidase/arabinofuranosidase gene (PcXyl) was cloned from the cDNA library of Phanerochaete chrysosporium and further expressed in Pichia pastoris. Enzymatic assay indicated that P. pastoris produced rPcXyl at a level of 26,141 U l⁻¹. The xylosidase and arabinofuranosidase activities of rPcXyl were maximized, respectively, at pHs of 5.0 and 5.5 and temperatures of 45°C and 50°C. SDS-PAGE revealed a single band of purified rPcXyl of 83 kDa. Cu²âº and Zn²âº completely inhibited the enzyme activity of rPcXyl. The enzyme activity of rPcXyl was increased 151%, 126% and 123%, respectively, in the presence of glucose, xylose and arabinose at concentrations of 5 mM. rPcXyl hydrolyzed xylobiose to xylose and xylotriose to xylose and xylobiose, indicating rPcXyl acts as an exo-type enzyme. Additionally, rPcXyl enhanced xylose release from xylan substrates in synergy with rPcXynC.


Subject(s)
Glycoside Hydrolases/metabolism , Phanerochaete/enzymology , Xylosidases/metabolism , Amino Acid Sequence , Arabinose/genetics , Cloning, Molecular , Disaccharides/metabolism , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/genetics , Molecular Sequence Data , Phanerochaete/genetics , Pichia/genetics , Recombinant Proteins/metabolism , Xylans/metabolism , Xylose/metabolism , Xylosidases/chemistry , Xylosidases/genetics
18.
Proc Natl Acad Sci U S A ; 107(7): 2890-5, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133652

ABSTRACT

A novel concept in eukaryotic signal transduction is the use of nutrient transporters and closely related proteins as nutrient sensors. The action mechanism of these "transceptors" is unclear. The Pho84 phosphate transceptor in yeast transports phosphate and mediates rapid phosphate activation of the protein kinase A (PKA) pathway during growth induction. We have now identified several phosphate-containing compounds that act as nontransported signaling agonists of Pho84. This indicates that signaling does not require complete transport of the substrate. For the nontransported agonist glycerol-3-phosphate (Gly3P), we show that it is transported by two other carriers, Git1 and Pho91, without triggering signaling. Gly3P is a competitive inhibitor of transport through Pho84, indicating direct interaction with its phosphate-binding site. We also identified phosphonoacetic acid as a competitive inhibitor of transport without agonist function for signaling. This indicates that binding of a compound into the phosphate-binding site of Pho84 is not enough to trigger signaling. Apparently, signaling requires a specific conformational change that may be part of, but does not require, the complete transport cycle. Using Substituted Cysteine Accessibility Method (SCAM) we identified Phe(160) in TMD IV and Val(392) in TMD VIII as residues exposed with their side chain into the phosphate-binding site of Pho84. Inhibition of both transport and signaling by covalent modification of Pho84(F160C) or Pho84(V392C) showed that the same binding site is used for transport of phosphate and for signaling with both phosphate and Gly3P. Our results provide to the best of our knowledge the first insight into the molecular mechanism of a phosphate transceptor.


Subject(s)
Proton-Phosphate Symporters/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Signal Transduction/physiology , Binding Sites/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Glycerophosphates/metabolism , Membrane Transport Proteins/metabolism , Mutagenesis, Site-Directed , Phosphonoacetic Acid/metabolism , Proton-Phosphate Symporters/agonists , Proton-Phosphate Symporters/genetics , Reproducibility of Results , Saccharomyces cerevisiae Proteins/agonists , Saccharomyces cerevisiae Proteins/genetics
19.
Acta Microbiol Immunol Hung ; 55(2): 75-89, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18595314

ABSTRACT

In yeast the Protein Kinase A (PKA) pathway can be activated by a variety of nutrients. Fermentable sugars, like glucose and sucrose, trigger a spike in the cAMP level, followed by activation of PKA and phosphorylation of target proteins causing a.o. mobilization of reserve carbohydrates, repression of stress-related genes and induction of growth-related genes. Glucose and sucrose are sensed by a G-protein coupled receptor system that activates adenylate cyclase and also activates a bypass pathway causing direct activation of PKA. Addition of other essential nutrients, like nitrogen sources or phosphate, to glucose-repressed nitrogen- or phosphate-starved cells, also triggers rapid activation of the PKA pathway. In these cases cAMP is not involved as a second messenger. Amino acids are sensed by the Gap1 transceptor, previously considered only as an amino acid transporter. Recent results indicate that the amino acid ligand has to induce a specific conformational change for signaling. The same amino acid binding site is involved in transport and signaling. Similar results have been obtained for Pho84 which acts as a transceptor for phosphate activation of the PKA pathway. Ammonium activation of the PKA pathway in nitrogen-starved cells is mediated mainly by the Mep2 transceptor, which belongs to a different class of transporter proteins. Hence, different types of sensing systems are involved in control of the yeast PKA pathway by nutrients.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Food , Saccharomyces/physiology , Signal Transduction , Fungal Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...