Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Omega ; 9(12): 14005-14016, 2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38559955

RESUMO

Helicobacter pylori infection is widespread in 50% of the world's population and is associated with gastric ulcers and related disorders that ultimately culminate in gastric cancer. Levofloxacin-based, or clarithromycin-based, triple therapy is frequently used to inhibit the bacterial urease enzyme for the eradication of H. pylori. A comprehensive investigation based on the urease inhibitory profiles of antibiotics and their computational implications is lacking in the scientific literature. The present study was aimed specifically to determine the antiurease activities within the realms of cephalosporins and fluoroquinolones by in vitro methods supported with in silico investigations. The results demonstrate the jack bean urease inhibitory activity of cephalosporins, wherein cefadroxil, cefpodoxime, cefotaxime, and cefaclor displayed inhibitions (IC50 21.35 ± 0.64 to 62.86 ± 0.78 µM) compared with the standard thiourea (IC50 21.25 ± 0.15 µM). Among fluoroquinolones, levofloxacin, ofloxacin, and gemifloxacin (IC50 7.24 ± 0.29 to 16.53 ± 0.85 µM) unveiled remarkable inhibitory profiles. Levofloxacin and ofloxacin exhibited competitive inhibition against the said enzyme. Ciprofloxacin and moxifloxacin displayed weak urease inhibitions. During molecular docking studies, Asp362, Gly279, Arg338, Asn168, Asp223, Gln364, and Met366 were involved in hydrogen bonding in fluoroquinolones, and hydrogen bonding was established with Arg338, His248, Asn168 residues, and metal Ni601 and Ni602 of the enzyme. MD simulations and MMPBSA results demonstrated the existence of significant protein-ligand binding. Overall, these results warrant further investigations into the significance of these active molecules in relation to their inhibitory potential against the targeted urease enzyme.

2.
Heliyon ; 10(2): e24470, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38298631

RESUMO

Montelukast, an approved leukotriene receptor 1 (Cys-LT 1) antagonist with anti-inflammatory properties is used for the treatment of asthma and allergic rhinitis. In the present studies, montelukast was subjected to in vitro inhibitory assays followed by kinetic and in silico investigations. Montelukast demonstrated inhibitory activity against yeast α-glucosidase (IC50 44.31 ± 1.21 µM), jack bean urease (JB urease, IC50 8.72 ± 0.23 µM), human placental alkaline phosphatase (hPAP, IC50 17.53 ± 0.19 µM), bovine intestinal alkaline phosphatase (bIAP, IC50 15.18 ± 0.23 µM) and soybean 15-lipoxygenase (15-LOX, IC50 2.41 ± 0.13 µM). Kinetic studies against α-glucosidase and urease enzymes revealed its competitive mode of inhibition. Molecular expression analysis of montelukast in breast cancer cell line MCF-7 down-regulated AP by a factor of 0.27 (5 µM) compared with the 0.26 value for standard inhibitor levamisole (10 µM). Molecular docking estimated a binding affinity ranging -8.82 to -15.65 kcal/mol for the enzymes. Docking against the DNA dodecamer (ID: 1BNA) observed -9.13 kcal/mol via minor groove binding. MD simulations suggested stable binding between montelukast and the target proteins predicting strong inhibitory potential of the ligand. Montelukast features a chloroquinoline, phenyl ring, a cyclopropane group, a carboxylic group and a sulfur atom all of which collectively enhance its inhibitory potential against the said enzymes. These in vitro and computational investigations demonstrate that it is possible and suggested that the interactions of montelukast with more than one targets presented herein may be linked with the side effects presented by this drug and necessitate additional work. The results altogether suggest montelukast as an important structural scaffold possessing multitargeted features and warrant further investigations in repurposing beyond its traditional pharmacological use.

3.
J Biomol Struct Dyn ; 42(6): 2913-2928, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37160706

RESUMO

The main goal of treating any Helicobacter pylori (H. pylori)-related gastrointestinal disease is completely eradicating infection. Falling eradication efficiency, off-target effects, and patient noncompliance with prolonged and broad spectrums have sparked clinical interest in exploring other effective, safer therapeutic choices. As natural substances are risk-free and privileged with high levels of antibacterial activity, most of these natural chemical's specific modes of action are unknown. With the aid of in silico molecular docking-based virtual screening studies and molecular dynamic simulations, the current study is intended to gather data on numerous such natural chemicals and assess their affinity for the S-ribosyl homocysteine lyase (LuxS) protein of H. pylori. The ligand with the highest binding energy with LuxS, glucoraphanin, catechin gallate and epigallocatechin gallate were rationally selected for further computational analysis. The solution stability of the three compounds' optimal docking postures with LuxS was initially assessed using long-run molecular dynamics simulations. Using molecular dynamics simulation, the epigallocatechin gallate was found to be the most stable molecule with the highest binding free energy, indicating that it might compete with the natural ligand of the inhibitors. According to ADMET analysis, his phytochemical was a promising therapeutic candidate for an antibacterial action since it had a range of physicochemical, pharmacokinetic, and drug-like qualities and had no discernible adverse effects. Additional in vitro, in vivo, and clinical trials are needed to confirm the drug's precise efficacy during H. pylori infection.Communicated by Ramaswamy H. Sarma.


Assuntos
Produtos Biológicos , Helicobacter pylori , Humanos , Simulação de Dinâmica Molecular , Ensaios de Triagem em Larga Escala , Simulação de Acoplamento Molecular , Ligantes , Produtos Biológicos/metabolismo , Resistência Microbiana a Medicamentos , Antibacterianos/farmacologia , Antibacterianos/metabolismo
4.
J Biomol Struct Dyn ; 42(7): 3616-3629, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37272194

RESUMO

The present study investigates the activity of the natural compound piperine on prostate cancer cell line (PC-3), followed by exploring its mechanistic inhibition on the RAC-alpha serine/threonine-protein kinase (AKT1) protein. The 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) assay showed that after 24 hrs of exposure to piperine (15 µmol/ml), cell viability fell to 50% compared to the standard drug flutamide (SDF) (51 µmol/ml) with a lower IC50 concentration. However, the Dual acridine orange/ethidium bromide (AO/EtBr) staining demonstrated that, as compared to the SDF, piperine caused substantial cellular death in PC-3 cells, presumably by triggering DNA fragmentation. In addition, compared to untreated cells, the proportion of the sub-G0/G1 and G2/M stages population increased considerably in piperine-treated cells. The cell cycle's sub-G0/G1 and G2/M phases were also arrested in piperine-treated cells compared to the SDF in cell cycle analysis. Based on our systems pharmacology and molecular docking studies, AKT1 is predicted as a potential target against piperine. The complementary charge between AKT1 and piperine was emphasized in the transient ligand-protein binding interaction in molecular dynamic modeling over 100 ns, and stable hydrogen bond interaction between Lys268 and Ser205 amino acid residues of the active pocket was hypothesized. Overall, the findings from our in vitro and MD simulations provide insights into the mechanism of piperine targeting AKT1 and offer a possible candidate for future prostate cancer therapeutic development.Communicated by Ramaswamy H. Sarma.


Assuntos
Alcaloides , Benzodioxóis , Piperidinas , Alcamidas Poli-Insaturadas , Neoplasias da Próstata , Masculino , Humanos , Simulação de Dinâmica Molecular , Simulação de Acoplamento Molecular , Neoplasias da Próstata/tratamento farmacológico , Alcaloides/farmacologia , Alcaloides/química , Proteínas Proto-Oncogênicas c-akt/metabolismo
5.
J Biomol Struct Dyn ; 42(6): 2765-2781, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37144749

RESUMO

Ferulic acid is a crucial bioactive component of broccoli, wheat, and rice bran and is also an essential natural product that has undergone significant research. Ferulic acid's precise mode of action and effect on system-level protein networks have not been thoroughly investigated. An interactome was built using the STRING database and Cytoscape tools, utilizing 788 key proteins collected from PubMed literature to identify the ferulic acid-governed regulatory action on protein interaction network (PIN). The scale-free biological network of ferulic acid-rewired PIN is highly interconnected. We discovered 15 sub-modules using the MCODE tool for sub-modulization analysis and 153 enriched signaling pathways. Further, functional enrichment of top bottleneck proteins revealed the FoxO signaling pathway involved in enhancing cellular defense against oxidative stress. The selection of the critical regulatory proteins of the ferulic acid-rewired PIN was completed by performing analyses of topological characteristics such as GO term/pathways analysis, degree, bottleneck, molecular docking, and dynamics investigations. The current research derives a precise molecular mechanism for ferulic acid's action on the body. This in-depth in silico model would aid in understanding how ferulic acid origins its antioxidant and scavenging properties in the human body.Communicated by Ramaswamy H. Sarma.


Assuntos
Ácidos Cumáricos , Simulação de Dinâmica Molecular , Mapas de Interação de Proteínas , Humanos , Simulação de Acoplamento Molecular , Ciclo-Oxigenase 2
6.
J Biomol Struct Dyn ; 41(17): 8472-8484, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36307909

RESUMO

This study aims to investigate the mechanism of natural antioxidant ferulic acid (FA) in reducing oxidative stress followed by its inhibitory effect on the Keap1-Nrf2 complex. FA was treated ex vivo with human blood for 30 min at 37 °C ± 1 °C and exposed to 1.5 Gy of γ- rays of 60Co (0.789 Gy/min) and allowed for repair for an hour at 37 °C ± 1 °C. FA's free radical scavenging capacity was measured using 2,7-dichlorofluorescein diacetate assay and cytogenetic assays. Further, a possible mechanism of protein-ligand interaction between FA and Keap1-Nrf2 pathway protein as a cellular drug target was studied using docking and molecular dynamics simulation. The 1.5 Gy of γ- rays exposed to pre-treated blood with FA showed a significant (p < 0.05) reduction in reactive oxygen species and DNA damage compared to the normal control blood group sample. The ligand-protein transient binding interaction in molecular dynamic simulation over a period of 100 ns was consistent and stable emphasizing complementary charge between the protein and ligand, speculating higher hydrophobic amino acid residues in the Keap1 active pocket. This might sway the Keap1 from interaction with Nrf2, and could lead to nuclear translocation of Nrf2 during radiation-induced oxidative stress. The present study emphasizes the radioprotective effect of FA against 1.5 Gy of γ- rays exposed to human blood and the application of in silico approaches helpful for the possible protective effect of FA.Communicated by Ramaswamy H. Sarma.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...