Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Heliyon ; 7(12): e08482, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34934830

RESUMO

ATP synthase subunit c (AtpE) is an enzyme that catalyzes the production of ATP from ADP in the presence of sodium or proton gradient from Mycobacterium tuberculosis (MTB). This enzyme considered an essential target for drug design and shares the same pathway with the target of Isoniazid. Thus, this enzyme would serve as an alternative target of the Isoniazid. The three dimensional (3D) model structure of the AtpE was constructed based on the principle of homology modeling using the Modeller9.16. The developed model was subjected to energy minimization and refinement using molecular dynamic (MD) simulation. The minimized model structure was searched against Zinc and PubChem database to determine ligands that bind to the enzyme with minimum binding energy using RASPD and PyRx tool. A total of 4776 compounds capable of bindings to AtpE with minimum binding energy were selected. These compounds further screened for physicochemical properties (Lipinski rule of five). All the compounds that possessed the desirable property selected and used for molecular docking analysis. Five (5) compounds with minimum binding energies ranged between ─8.69, and ─8.44 kcal/mol, less than the free binding energy of ATP were selected. These compounds further screened for the absorption, distribution, metabolism, excretion, and toxicity (ADME and toxicity) properties. Of the five compounds, three (ZINC14732869, ZINC14742188, and ZINC12205447) fitted all the ADME and toxicity properties and subjected to MD simulation and Molecular Mechanics Generalized Born and Surface Area (MM-GBSA) analyses. The results indicated that the ligands formed relatively stable complexes and had free binding energies, less than the binding energy of the ATP. Therefore, these ligands considered as prospective inhibitors of MTB after successful experimental validation.

2.
Life Sci ; 262: 118466, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32961233

RESUMO

INTRODUCTION: UDP-N-acetylmuramic-alanine ligase (MurC) is an enzyme catalyzing the addition of L-alanine to UDP-acetylmuramoyl nucleotide precursor in Mycobacterium tuberculosis (M. tuberculosis). This enzyme is a prerequisite for the biosynthesis of the peptidoglycans in M. tuberculosis. AIM: This study aimed to identify the novel inhibitors of MurC using in silico approach. MATERIALS AND METHODS: The three dimensional (3D) structure of MurC was determined using comparative modeling and based on the template obtained from Haemophilus influenza (1P31). The structural analysis of the model structure shown that three residues (Lys126, Glu170, and Glu358) are critical for in the catalytic activity of the enzyme, and their inhibition will block the function of the enzyme. Ten thousand and ninety-five (10095) compounds obtained through virtual screening against Zinc and PubChem databases based on their ability to bind to MurC with minimum binding energies. These ligands screened for the physicochemical properties, molecular docking, and pharmacokinetic analyses. FINDING: Six compounds had desirable physicochemical and pharmacokinetic properties with excellent binding energy ranged between -12.27 and -10.09 kcal/mol. These compounds subjected to Molecular Dynamic (MD) Simulation and Molecular Mechanics Generalized Born Surface Area (MM-GBSA) analyses. The outcome of the analysis revealed that four ligands (PubChem1548994, ZINC11882115, ZINC22241774, and ZINC12330603) formed a stable conformation in the substrate-binding site of the protein during the 50 ns MD simulation. CONCLUSION: Therefore, the ligands mentioned above might regard as novel inhibitors of M. tuberculosis which requires further in vitro and in vivo validation.


Assuntos
Antituberculosos/farmacologia , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/efeitos dos fármacos , Peptídeo Sintases/antagonistas & inibidores , Antituberculosos/química , Antituberculosos/farmacocinética , Sítios de Ligação , Ligantes , Simulação de Acoplamento Molecular , Mycobacterium tuberculosis/enzimologia , Peptídeo Sintases/química , Ligação Proteica
3.
Int J Biol Macromol ; 160: 623-631, 2020 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-32473219

RESUMO

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.


Assuntos
Glutationa/farmacologia , Lipase/antagonistas & inibidores , Pâncreas/efeitos dos fármacos , Aminoácidos/metabolismo , Animais , Catálise/efeitos dos fármacos , Humanos , Cinética , Simulação de Acoplamento Molecular/métodos , Simulação de Dinâmica Molecular , Espectrometria de Fluorescência/métodos , Suínos
4.
Int. microbiol ; 22(1): 7-17, mar. 2019. graf, tab
Artigo em Inglês | IBECS | ID: ibc-184809

RESUMO

The present study attempts to identify the novel inhibitors of shikimate dehydrogenase (SD), the enzyme that catalyzes the fourth reaction in the shikimate pathway, through virtual screening and toxicity studies. Crystal structure of SD was obtained from Protein Data Bank (PDB ID 4P4G, 1.7 Å) and subjected to energy minimization and structure optimization. A total of 13,803 compounds retrieved from two public databases and used for the virtual screening based on physicochemical properties (Lipinski rule of five) and molecular docking analyses. A total of 26 compounds with good AutoDock binding energies values ranging between −12.03 and −8.33 kcal/mol was selected and further filtered for absorption distribution metabolism excretion and toxicity analyses (ADMET). In this, eight compounds were selected, which satisfied all the ADME and toxicity analysis properties. Three compounds with better AutoDock binding energies values (ZINC12135132, −12.03 kcal/mol; ZINC08951370, −10.04 kcal/mol; and ZINC14733847, 9.82 kcal/mol) were considered for molecular dynamic (MD) simulation and molecular generalized born surface area (MM-GBSA) analyses. The results of the analyses revealed that the two ligands (ZINC12135132 and ZINC08951370) had better inhibitory activities within their complexes, after the 50-ns MD simulation, which suggested that the complexes formed stable conformation. It is noteworthy that compounds identified by docking, MD simulation, and MM-GBSA methods could be a drug for tuberculosis which required further experimental validation


No disponible


Assuntos
Oxirredutases/administração & dosagem , Tuberculose/tratamento farmacológico , Oxirredutases do Álcool/antagonistas & inibidores , Antituberculosos/isolamento & purificação , Biologia Computacional/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/isolamento & purificação , Tuberculose/microbiologia , Oxirredutases do Álcool/química , Antituberculosos/toxicidade , Inibidores Enzimáticos/toxicidade , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica
5.
Int Microbiol ; 22(1): 7-17, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30810932

RESUMO

The present study attempts to identify the novel inhibitors of shikimate dehydrogenase (SD), the enzyme that catalyzes the fourth reaction in the shikimate pathway, through virtual screening and toxicity studies. Crystal structure of SD was obtained from Protein Data Bank (PDB ID 4P4G, 1.7 Å) and subjected to energy minimization and structure optimization. A total of 13,803 compounds retrieved from two public databases and used for the virtual screening based on physicochemical properties (Lipinski rule of five) and molecular docking analyses. A total of 26 compounds with good AutoDock binding energies values ranging between - 12.03 and - 8.33 kcal/mol was selected and further filtered for absorption distribution metabolism excretion and toxicity analyses (ADMET). In this, eight compounds were selected, which satisfied all the ADME and toxicity analysis properties. Three compounds with better AutoDock binding energies values (ZINC12135132, - 12.03 kcal/mol; ZINC08951370, - 10.04 kcal/mol; and ZINC14733847, 9.82 kcal/mol) were considered for molecular dynamic (MD) simulation and molecular generalized born surface area (MM-GBSA) analyses. The results of the analyses revealed that the two ligands (ZINC12135132 and ZINC08951370) had better inhibitory activities within their complexes, after the 50-ns MD simulation, which suggested that the complexes formed stable conformation. It is noteworthy that compounds identified by docking, MD simulation, and MM-GBSA methods could be a drug for tuberculosis which required further experimental validation.


Assuntos
Oxirredutases do Álcool/antagonistas & inibidores , Antituberculosos/isolamento & purificação , Biologia Computacional/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Inibidores Enzimáticos/isolamento & purificação , Oxirredutases do Álcool/química , Antituberculosos/química , Antituberculosos/farmacologia , Antituberculosos/toxicidade , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/toxicidade , Simulação de Acoplamento Molecular , Ligação Proteica , Conformação Proteica , Tuberculose/tratamento farmacológico
6.
Comput Biol Chem ; 78: 116-126, 2019 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-30504089

RESUMO

The present study aimed to identify the prospective inhibitors of MurD, a cytoplasmic enzyme that catalyzes the addition of d-glutamate to the UDP-N-acetylmuramoyl-l-alanine nucleotide precursor in Mycobacterium tuberculosis (MTB), using virtual screening, docking studies, pharmacokinetic analysis, Molecular Dynamic (MD) simulation, and Molecular Mechanics Generalized Born and Surface Area (MM-GBSA) analyses. The three dimensional (3D) structure was determined based on the homology technique using a template from Streptococcus agalactiae. The modeled structure had three binding sites, namely; substrate binding site (Val18, Thr19, Asp39, Asp40, Gly75, Asn147, Gln171 and His192), the ATP binding site (Gly123, Lys124, Thr125, Thr126, Glu166, Asp283, and Arg314) and the glutamic acid binding site (Arg382, Ser463, and Tyr470). These residues mentioned above play a critical role in the catalytic activity of the enzyme, and their inhibition could serve as a stumbling block to the normal function of the enzyme. A total of 10,344 obtained from virtual screened of Zinc and PubChem databases. These compounds further screened for Lipinski rule of five, docking studies and pharmacokinetic analysis. Four compounds with good binding energies (ZINC11881196 = -10.33 kcal/mol, ZINC12247644 = -8.90 kcal/mol, ZINC14995379 =-8.42 kcal/mol, and PubChem6185 = -8.20 kcal/mol), better than the binding energies of the ATP (-2.31 kcal/mol) and the ligand with known IC50, Aminothiazole (-7.11 kcal/mol) were selected for the MD simulation and MM-GBSA analyses. The result of the analyses showed that all the four ligands formed a stable complex and had the binding free energies better than the binding energy of ATP. Therefore, these ligands considered as suitable prospective inhibitors of the MurD after experimental validation.


Assuntos
Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/enzimologia , Peptídeo Sintases/química , Ligantes , Estrutura Molecular , Peptídeo Sintases/antagonistas & inibidores , Peptídeo Sintases/metabolismo , Propriedades de Superfície , Termodinâmica
7.
J Mol Model ; 24(6): 132, 2018 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-29752576

RESUMO

The shikimate pathway is as an attractive target because it is present in bacteria, algae, fungi, and plants but does not occur in mammals. In Mycobacterium tuberculosis (MTB), the shikimate pathway is integral to the biosynthesis of naphthoquinones, menaquinones, and mycobactin. In these study, novel inhibitors of 3-dehydroquinate synthase (DHQS), an enzyme that catalyzes the second step of the shikimate pathway in MTB, were determined. 12,165 compounds were selected from two public databases through virtual screening and molecular docking analysis using PyRx 8.0 and Autodock 4.2, respectively. A total of 18 compounds with the best binding energies (-13.23 to -8.22 kcal/mol) were then selected and screened for absorption, distribution, metabolism, excretion, and toxicity (ADMET) analysis, and nine of those compounds were found to satisfy all of the ADME and toxicity criteria. Among those nine, the three compounds-ZINC633887 (binding energy = -10.29 kcal/mol), ZINC08983432 (-9.34 kcal/mol), and PubChem73393 (-8.61 kcal/mol)-with the best binding energies were further selected for molecular dynamics (MD) simulation analysis. The results of the 50-ns MD simulations showed that the two compounds ZINC633887 and PubChem73393 formed stable complexes with DHQS and that the structures of those two ligands remained largely unchanged at the ligand-binding site during the simulations. These two compounds identified through docking and MD simulation are potential candidates for the treatment of TB, and should undergo validation in vivo and in vitro.


Assuntos
Proteínas de Bactérias/química , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Mycobacterium tuberculosis/enzimologia , Fósforo-Oxigênio Liases/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...