Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Bioorg Chem ; 147: 107361, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613924

ABSTRACT

Biologically important macromolecule 1, 1', 3, 3' Bis - [2,3,5,6-Tetramethyl-p-phenylenebis(methylene)] dibenzotriazlinium dibromide hydrate (BTD) was synthesized and characterized using FT-IR, NMR and single-crystal XRD (SCXRD). SCXRD revealed that the compound was crystallized as a monoclinic system and associated through weak intermolecular interactions like H-bonding and π- π stacking interactions. These weak intermolecular interactions in BTD were studied using Crystal Explorer and Gaussian. The calculated energies for the Highest Occupied Molecular Orbital (HOMO) and the Lowest Unoccupied Molecular Orbital (LUMO) showed the stability and reactivity of the title compound. Molecular electrostatic potential (MEP) surface analysis was used to investigate the crystal's nucleophilic and electrophilic reactive sites. The molecular shape and intermolecular interactions in the crystal structure were determined using Hirshfeld surface analysis and fingerprint plots. Anticancer, anti-bacterial and DNA binding ability of BTD were investigated by experimental and theoretical techniques. The obtained results suggest that BTD possesses better anti-cancer, anti-bacterial and DNA binding abilities. The mode of action of antibiotic and anticancer approach was discussed. This provides promising therapeutic advantages for further development.


Subject(s)
Antineoplastic Agents , Antitubercular Agents , DNA , Molecular Docking Simulation , Triazoles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Triazoles/chemistry , Triazoles/pharmacology , Triazoles/chemical synthesis , Humans , Ligands , Antitubercular Agents/pharmacology , Antitubercular Agents/chemistry , Antitubercular Agents/chemical synthesis , Molecular Structure , DNA/chemistry , DNA/metabolism , Structure-Activity Relationship , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/pharmacology , Macrocyclic Compounds/chemical synthesis , Microbial Sensitivity Tests , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Mycobacterium tuberculosis/drug effects , Cell Proliferation/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis
2.
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
3.
Appl Biochem Biotechnol ; 193(6): 1909-1923, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33512650

ABSTRACT

The unprecedented coronavirus SARS-CoV-2 outbreak at Wuhan, China, caused acute respiratory infection to humans. There is no precise vaccine/therapeutic agents available to combat the COVID-19 disease. Some repurposed drugs are saving the life of diseased, but the complete cure is relatively less. Several drug targets have been reported to inhibit the SARS-CoV-2 virus infection, in that TMPRSS2 (transmembrane protease serine 2) is one of the potential targets; inhibiting this protease stops the virus entry into the host human cell. Camostat mesylate, nafamostat, and leupeptin are the drugs, in which the first two drugs are being used for COVID-19 and leupeptin also tested. To consider these drugs as the repurposed drug for COVID-19, it is essential to understand their binding affinity and stability with TMPRSS2. In the present study, we performed the molecular docking and molecular dynamics (MD) simulation of these molecules with the TMPRSS2. The docking study reveals that leupeptin molecule strongly binds with TMPRSS2 protein than the other two drug molecules. The RMSD and RMSF values of MD simulation confirm that leupeptin and the amino acids of TMPRSS2 are very stable than the other two molecules. Furthermore, leupeptin forms interactions with the key amino acids of TMPRSS2 and the same have been maintained during the MD simulations. This structural and dynamical information is useful to evaluate these drugs to be used as repurposed drugs, however, the strong binding profile of leupeptin with TMPRSS2, suggests, it may be considered as a repurposed drug for COVID-19 disease after clinical trial.


Subject(s)
Antiviral Agents/pharmacology , Benzamidines/therapeutic use , COVID-19 Drug Treatment , Drug Repositioning , Esters/therapeutic use , Guanidines/therapeutic use , Leupeptins/metabolism , Molecular Docking Simulation , Molecular Dynamics Simulation , Serine Endopeptidases/metabolism , Antiviral Agents/therapeutic use , Benzamidines/pharmacology , COVID-19/virology , Esters/pharmacology , Guanidines/pharmacology , Humans , Protein Binding , SARS-CoV-2/drug effects
4.
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
5.
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
6.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...