Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros










Intervalo de año de publicación
1.
Mol Divers ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418686

RESUMEN

In this study, we explored the potential of novel inhibitors for FYN kinase, a critical target in cancer and neurodegenerative disorders, by integrating advanced cheminformatics, machine learning, and molecular simulation techniques. Our approach involved analyzing key interactions for FYN inhibition using established multi-kinase inhibitors such as Staurosporine, Dasatinib, and Saracatinib. We utilized ECFP4 circular fingerprints and the t-SNE machine learning algorithm to compare molecular similarities between FDA-approved drugs and known clinical trial inhibitors. This led to the identification of potential inhibitors, including Afatinib, Copanlisib, and Vandetanib. Using the DrugSpaceX platform, we generated a vast library of 72,196 analogues from these leads, which after careful refinement, resulted in 6008 promising candidates. Subsequent clustering identified 48 analogues with significant similarity to known inhibitors. Notably, two candidates derived from Vandetanib, DE27123047 and DE27123035, exhibited strong docking affinities and stable binding in molecular dynamics simulations. These candidates showed high potential as effective FYN kinase inhibitors, as evidenced by MMGBSA calculations and MCE-18 scores exceeding 50. Additionally, our exploration into their molecular architecture revealed potential modification sites on the quinazolin-4-amine scaffold, suggesting opportunities for strategic alterations to enhance activity and optimize ADME properties. Our research is a pioneering effort in drug discovery, unveiling novel candidates for FYN inhibition and demonstrating the efficacy of a multi-layered computational strategy. The molecular insights gained provide a pathway for strategic refinements and future experimental validations, setting a new direction in targeted drug development against diseases involving FYN kinase.

2.
Sci Rep ; 14(1): 3872, 2024 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-38365839

RESUMEN

Hemigraphis alternata (H. alternata), commonly known as Red Flame Ivy, is widely recognized for its wound healing capabilities. However, the pharmacologically active plant components and their mechanisms of action in wound healing are yet to be determined. This study presents the mass spectrometry-based global metabolite profiling of aqueous and ethanolic extract of H. alternata leaves. The analysis identified 2285 metabolites from 24,203 spectra obtained in both positive and negative polarities. The identified metabolites were classified under ketones, carboxylic acids, primary aliphatic amines, steroids and steroid derivatives. We performed network pharmacology analysis to explore metabolite-protein interactions and identified 124 human proteins as targets for H. alternata metabolites. Among these, several of them were implicated in wound healing including prothrombin (F2), alpha-2A adrenergic receptor (ADRA2A) and fibroblast growth factor receptor 1 (FGFR1). Gene ontology analysis of target proteins enriched cellular functions related to glucose metabolic process, platelet activation, membrane organization and response to wounding. Additionally, pathway enrichment analysis revealed potential molecular network involved in wound healing. Moreover, in-silico docking analysis showed strong binding energy between H. alternata metabolites with identified protein targets (F2 and PTPN11). Furthermore, the key metabolites involved in wound healing were further validated by multiple reaction monitoring-based targeted analysis.


Asunto(s)
Activación Plaquetaria , Cicatrización de Heridas , Humanos , Cicatrización de Heridas/fisiología , Metabolómica , Hojas de la Planta/química , Simulación del Acoplamiento Molecular
3.
J Biomol Struct Dyn ; : 1-13, 2024 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-38239070

RESUMEN

In the era of targeted therapeutics, protein kinases like WEE1 have become pivotal drug targets, especially for cancer therapy. Utilizing a multi-faceted approach, our study adds fresh insights to this endeavour. We employed the t-SNE algorithm, combined with ECFP4 fingerprints, to analyse the molecular similarity between FDA-approved drugs and known clinical trial inhibitors. Our t-SNE analysis identified the closest clusters to known inhibitors and selected 11 FDA-approved drugs for further study. Using the DrugSpaceX platform, we generated analogues for these 11 FDA-approved drugs. These analogues were refined according to Lipinski's Rule of Five and Synthetic Accessibility scores, yielding 68,640 analogues for additional scrutiny. Among these, derivatives of Palbociclib and Ribociclib stood out as the most promising WEE1 inhibitors, based on docking scores and interaction patterns. Molecular dynamics simulations validated the stability of these protein-ligand interactions, particularly for DE50607359, a top-ranked Palbociclib analogue, which also met most pharmacokinetic parameters within acceptable limits. Our study uncovers new candidates for WEE1 inhibition not previously reported. With our multi-layered computational strategy, we provide a solid foundation for future experimental validation and targeted drug development in cancer therapeutics.Communicated by Ramaswamy H. Sarma.


Employed the t-SNE algorithm and ECFP4 fingerprints to discern molecular similarities between FDA-approved drugs and known clinical trial inhibitors, identifying 11 key drugs.Leveraged the DrugSpaceX platform to generate analogues for these selected FDA-approved drugs, yielding a massive collection of 68,640 refined analogues based on Lipinski's Rule of Five and Synthetic Accessibility scores.Derivatives of Palbociclib and Ribociclib emerged as the most promising WEE1 inhibitors, supported by their docking scores and interaction patterns.Validated protein-ligand interactions through molecular dynamics simulations, spotlighting DE50607359, a superior Palbociclib analogue, meeting critical pharmacokinetic parameters.

4.
OMICS ; 28(1): 8-23, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38190280

RESUMEN

Checkpoint kinase 1 (CHK1), a serine/threonine kinase, plays a crucial role in cell cycle arrest and is a promising therapeutic target for drug development against cancers. CHK1 coordinates cell cycle checkpoints in response to DNA damage, facilitating repair of single-strand breaks, and maintains the genome integrity in response to replication stress. In this study, we employed an integrated computational and experimental approach to drug discovery and repurposing, aiming to identify a potent CHK1 inhibitor among existing drugs. An e-pharmacophore model was developed based on the three-dimensional crystal structure of the CHK1 protein in complex with CCT245737. This model, characterized by seven key molecular features, guided the screening of a library of drugs through molecular docking. The top 10% of scored ligands were further examined, with procaterol emerging as the leading candidate. Procaterol demonstrated interaction patterns with the CHK1 active site similar to CHK1 inhibitor (CCT245737), as shown by molecular dynamics analysis. Subsequent in vitro assays, including cell proliferation, colony formation, and cell cycle analysis, were conducted on gastric adenocarcinoma cells treated with procaterol, both as a monotherapy and in combination with cisplatin. Procaterol, in synergy with cisplatin, significantly inhibited cell growth, suggesting a potentiated therapeutic effect. Thus, we propose the combined application of cisplatin and procaterol as a novel potential therapeutic strategy against human gastric cancer. The findings also highlight the relevance of CHK1 kinase as a drug target for enhancing the sensitivity of cytotoxic agents in cancer.


Asunto(s)
4-Aminopiridina/análogos & derivados , Antineoplásicos , Pirazinas , Neoplasias Gástricas , Humanos , Cisplatino/farmacología , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Procaterol , Neoplasias Gástricas/tratamiento farmacológico , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Reposicionamiento de Medicamentos , Simulación del Acoplamiento Molecular , Línea Celular Tumoral , Antineoplásicos/farmacología , Descubrimiento de Drogas , Daño del ADN , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química
5.
Folia Morphol (Warsz) ; 83(1): 72-82, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-36967622

RESUMEN

BACKGROUND: Human umbilical cord (hUC) is encompassed by a mucoid connective tissue called Wharton's jelly (WJ), made of hyaluronic acid, collagen, and stromal cells to support the blood vessels of hUC. This study was aimed to determine the in vitro neuronal differentiation of WJ-derived mesenchymal stem cells (WJMSCs), and in vivo axonal regeneration potential of nanofiber coated human Wharton's jelly as a neuronal graft after sciatic nerve injury in immunosuppressed albino Wistar rats. MATERIALS AND METHODS: Wharton's jelly-derived mesenchymal stem cells could be differentiated to neuron-like cells by inducing with neuronic supplementing media. The test animal's axotomized nerves were implanted with trimmed human umbilical cord devoid of vascularity and nanocoated with electro-spun poly-l-lactic acid nanofibers. The control animals were bridged with native sciatic nerve reversed and sutured. Post-surgical functional recovery was studied by walking track, pinprick, muscle weight, and sweating quantification. At the end of the 4th week, the animals were euthanized, and magnetoneurography was performed. The explanted grafts were quantified by immunohistochemistry for immuno-rejection, neural scarring, neural adhesion axon regeneration, fibre diameter, myelin thickness, and G-ratio. The sciatic function index values were similar by walking track analysis for both the test and control groups. RESULTS: The animals had functional and sensation recovery by the end of 2 weeks. No mortality, signs of inflammation, and acute immune rejection were observed post-surgery. CONCLUSIONS: The hUCWJ devoid of vascular elements can be a perfect peripheral nerve graft, and we hypothesis that the cryopreserved hUC could be an ideal resource for axonal regeneration in the future.


Asunto(s)
Nanofibras , Ratas , Animales , Humanos , Ratas Wistar , Axones , Regeneración Nerviosa , Cordón Umbilical , Nervio Ciático/cirugía
6.
J Biomol Struct Dyn ; : 1-11, 2023 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-37632319

RESUMEN

The protein kinase Wee1 plays a vital role in the G2/M cell cycle checkpoint activation, triggered by double-stranded DNA disruptions. It fulfills this task by phosphorylating and consequently deactivating the cyclin B linked to Cdk1/Cdc2 at the Tyr15 residue, leading to a G2 cell cycle halt and subsequent delay of mitosis post DNA damage. Despite advancements, only the Wee1 inhibitor MK1775 has made it to Phase II clinical trials, presenting a challenge in innovative chemical structure development for small molecule discovery. To navigate this challenge, we employed an e-pharmacophore model of the MK1775-WEE1 complex (PDB ID: 5V5Y), using in silico screening of FDA-approved drugs. We chose six drugs for analog creation, guided by docking scores, key residue interactions, and ligand occupancy. Utilizing the 'DrugSpaceX' database, we generated 2,776 analogues via expert-defined transformations. Our findings identified DE90612 as the top-ranked analogue, followed by DE363106, DE489678, DE395383, DE90548, DE689343, DE395019, and DE538066. These analogues introduced unique structures not found in other databases. A t-SNE structurally diversified distribution map unveiled promising transformations linked to Temozolomide for WEE1 inhibitor development. Simulations of the WEE1-DE90612 complex (a Temozolomide analogue) for 200 nanoseconds demonstrated stability, with DE90612 forging robust bonds with active site residues and sustaining vital contacts at ASN376 and CYS379. These results underscore DE90612's potential inhibitory properties at the WEE1 binding site, warranting additional in vitro and in vivo exploration for its anticancer activity. Our approach outlines a promising pathway for creating diverse WEE1 inhibitors with suitable biological properties for potential oncology therapeutics.Communicated by Ramaswamy H. Sarma.

7.
Toxicol Mech Methods ; 33(9): 707-718, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37455591

RESUMEN

INTRODUCTION: C-Glucosyl Xanthone derivatives were assessed to inhibit the JNK3 mediated Caspase pathway in Almal (Aluminum Maltolate) induced neurotoxicity in SHSY-5Y cells. METHODS: Mangiferin was selected among 200 C-Glucosyl Xanthones based on molecular interaction, docking score (-10.22 kcal/mol), binding free energy (-71.12 kcal/mol), ADME/tox properties and by molecular dynamic studies. Further, it was noticed that glycone moiety of Mangiferin forms H-bond with ASN 194, SER 193, GLY 76, and OH group in the first position of the aglycone moiety shows interaction at Met 149 which is exceptionally crucial for JNK3 inhibitory activity. RESULTS AND DISCUSSION: Mangiferin (0.5, 1, 10, 20 and 30 µM) and standard SP600125 (20 µM) treatment increased the cell survival rate against Almal 200 µM, with EC50 of Mangiferin (8 µM) and standard SP600125 (4.9 µM) respectively. Mangiferin significantly impedes kinase activation, indicating suppression of JNK3 signaling with IC50 (98.26 nM). Mangiferin (10 and 15 µM) dose-dependently inhibits the caspase 3, 8, and 9 enzyme activation in comparison to Almal group. CONCLUSION: Mangiferin demonstrated neuroprotection in SHSY-5Y cells against apoptosis induced by Almal by adapting the architecture of the neurons and increasing their density. Among all Xanthone derivatives, Mangiferin could improve neuronal toxicity by inhibiting JNK3 and down-regulating the Caspase activation.


Asunto(s)
Neuroblastoma , Xantonas , Humanos , Xantonas/farmacología , Xantonas/química , Xantonas/metabolismo , Caspasas
8.
J Biomol Struct Dyn ; 41(24): 15196-15206, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37029757

RESUMEN

The calcium/calmodulin dependent protein kinase kinase 2 (CAMKK2) plays a key role in regulation of intracellular calcium levels and signaling pathways. It is involved in activation of downstream signaling pathways that regulate various cellular processes. Dysregulation of CAMKK2 activity has been linked to various diseases including cancer, suggesting that CAMKK2 inhibitors might be beneficial in oncological, metabolic and inflammatory indications. The most pressing issues in small molecule discovery are synthesis feasibility, novel chemical structure and desired biological characteristics. To circumvent this constraint, we employed 'DrugspaceX' for rapid lead identification, followed by repositioning seven FDA-approved drugs for CAMKK2 inhibition. Further, first-level transformation (Set1 analogues) was performed in 'DrugspaceX', followed by virtual screening. The t-SNE visualization revealed that the transformations surrounding Rucaparib, Treprostinil and Canagliflozin are more promising for developing CAMKK2 inhibitors. Second, using the top-ranked Set1 analogues, Set2 analogues were generated, and virtual screening revealed the top-ranked five analogues. Among the top five Set2 analogues, DE273038_5 had the lowest docking score of -11.034 kcal/mol and SA score of 2.59, retaining the essential interactions with Hotspot residues LYS194 and VAL270 across 250 ns simulation period. When compared to the other four compounds, the ligand effectiveness score was 0.409, and the number of rotatable penalties was only three. Further, DE273038_5 after two rounds of transformations was discovered to be novel and had not been previously described in other databases. These data suggest that the new candidate DE273038_5 is likely to have inhibitory activity at the CAMKK2 active site, implying potential therapeutic use.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina , Calcio , Calcio/metabolismo , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/química , Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Dominio Catalítico , Transducción de Señal
9.
Mol Divers ; 27(4): 1633-1644, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36006501

RESUMEN

Drug repurposing opens new avenues in cancer therapy. Drug repurposing, or finding new uses for existing drugs, can substantially reduce drug discovery time and costs. Cheminformatics, genetics, and systems biology advances enable repositioning drugs. Clinical usage of PD-1/PD-L1 blocking has been approved because of its efficacy in improving prognosis in select groups. The PD-1/PD-L1 axis was considered to represent a mechanism for tumour evasion of host tumour antigen-specific T-cell immunity in early preclinical research. The expression of PD-L1 in cancer cells causes T lymphocytes to become exhausted by transmitting a co-inhibitory signal. A better understanding of how PD-L1 is regulated in cancer cells could lead to new therapeutic options. In this view, the study was aimed to repurpose the existing FDA-approved drugs as a potential PD-L1 inhibitor through e-Pharmacophore modelling, molecular docking and dynamic simulation. e-Pharmacophore screening retrieved 324 FDA-approved medications with the fitness score ≥ 1. The top 10-docked FDA candidates were compared with IN-35 (Clinical trial candidate) for its interaction pattern with critical amino acid residues. Mirabegron and Indacaterol exhibited a greater affinity for PD-L1 with docking scores of - 9.213 kcal mol-1 and - 8.023 kcal mol-1, respectively. Mirabegron retain interactions at all three major hotspots in the PD-L1 dimer interface similar to IN-35. MM-GBSA analyses indicated that Mirabegron uses less energy to create a more stable complex and retains all of the inhibitor's positive interactions found in clinical trial ligand IN-35. Molecular dynamics simulation analysis of the Mirabegron complex showed a similar pattern of deviation in correlation with IN-35, and it retains the interaction with the active key amino acids throughout the simulation time. Our present study has shown Mirabegron as a powerful inhibitor of PD-L1 expression in cancer cells using a drug-repurposing screen.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Simulación del Acoplamiento Molecular , Antígeno B7-H1/química , Receptor de Muerte Celular Programada 1 , Simulación de Dinámica Molecular , Aminoácidos , Neoplasias/tratamiento farmacológico
10.
Int. microbiol ; 25(3): 629-638, Ago. 2022. ilus
Artículo en Inglés | IBECS | ID: ibc-216220

RESUMEN

Biofilm formation associated with quorum sensing (QS) is a community behaviour displayed by many gram-negative pathogenic bacteria that provide survival advantages in hostile conditions. The inhibitors of QS interrupt bacterial communication and coordinated cell signalling for community aggregation in the biofilm. Thymol, a natural monoterpenoid, was tested against QS in Chromobacterium violaceum. As the first step, the interaction of thymol with cviR protein was investigated using in silico approach followed by validation using detailed in vitro experiments. The QS and biofilm studies were performed using the wild type of strain C. violaceum ATCC 12,472 and a mini-Tn5 mutant CV026. The MIC of thymol was established by the broth micro-dilution method, and IC50 value for violacein inhibition was quantified spectrophotometrically by extracting the violacein from the treated cells. Inhibitory effect of thymol on the biofilm was quantified by the crystal violet staining method, and scanning electron microscopy (SEM) was employed for biofilm visualization. The expression of biofilm associated genes (hmsH, hmsR, pilB, and pilT) was evaluated by qRT-PCR analysis. The in silico molecular interactions of thymol with cviR exhibited a G-score of − 5.847 kcal/mol, binding with TYR-80 and SER-155 by Pi-Pi stacking and H-bond, respectively. The MIC of thymol was 160 µg/mL, and the IC50 for violacein inhibition was estimated to be 28 µg/mL. The thymol treatment significantly reduced the biofilm viability and biomass by > 80% along with disruption of the well-organized biofilm architecture. QS inhibitory activity of thymol resulted in the reduction of exopolysaccharide production, swarming motility, and downregulation of biofilm-associated hmsH, hmsR, pilB, and pilT genes. This data establishes the QS inhibitory role of thymol in the biofilm formation in C. violaceum.(AU)


Asunto(s)
Humanos , Timol , Biopelículas , Chromobacterium , Percepción de Quorum , Bacterias Gramnegativas , Microbiología
11.
Struct Chem ; 33(5): 1391-1407, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35855326

RESUMEN

COVID-19 infection is associated with a significant fatality rate in individuals suffering from severe acute respiratory distress syndrome (ARDS). Among the several possibilities, inhibition of hypoxia-inducible factor prolyl hydroxylase-2 or prolyl hydroxylase domain-containing protein 2 (PHD2) in a hypoxia-independent way is a prospective therapeutic target for the treatment of ARDS. Vadadustat, Roxadustat, Daprodustat, Desidustat, and Enarudustat are the available clinical trial inhibitors. This study is proposed to focus on the repurposing of FDA-approved drugs as effective PHD2 inhibitors. This computational study utilises e-pharmacophore hypothesis generation from the native ligand-protein complex (PDB ID: 5OX6) based on XP visualiser information. The hypothesis containing five essential features (AAANR) was incorporated for FDA database screening, followed by Glide XP molecular docking and Prime MM-GBSA binding free energy calculations. Top scored ligands were investigated and Fenbufen was identified as an effective PHD-2 inhibitor by comparing with the native co-crystal ligand (Vadadustat). The manual lead optimisation of the Fenbufen structure was adopted to improve inhibitory potency, by increasing the binding affinity and protein-ligand stability. The newly designed compounds B and C showed additional binding interactions, excellent docking scores, binding free energy, and an acceptable range of ADME properties. Also, Fenbufen and compound C owned preferable protein-ligand stability during MD simulation when compared with the co-crystallised clinical trial ligand. Based on our findings, we deduce that Fenbufen can be proposed as an effective repurposable candidate as its structural modification showed a remarkable improvement in PHD2 inhibition. Supplementary information: The online version contains supplementary material available at 10.1007/s11224-022-02012-z.

12.
Molecules ; 27(12)2022 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-35744923

RESUMEN

Plants and their derived molecules have been traditionally used to manage numerous pathological complications, including male erectile dysfunction (ED). Mimosa pudica Linn. commonly referred to as the touch-me-not plant, and its extract are important sources of new lead molecules in drug discovery research. The main goal of this study was to predict highly effective molecules from M. pudica Linn. for reaching and maintaining penile erection before and during sexual intercourse through in silico molecular docking and dynamics simulation tools. A total of 28 bioactive molecules were identified from this target plant through public repositories, and their chemical structures were drawn using Chemsketch software. Graph theoretical network principles were applied to identify the ideal target (phosphodiesterase type 5) and rebuild the network to visualize the responsible signaling genes, proteins, and enzymes. The 28 identified bioactive molecules were docked against the phosphodiesterase type 5 (PDE5) enzyme and compared with the standard PDE5 inhibitor (sildenafil). Pharmacokinetics (ADME), toxicity, and several physicochemical properties of bioactive molecules were assessed to confirm their drug-likeness property. Molecular dynamics (MD) simulation modeling was performed to investigate the stability of PDE5-ligand complexes. Four bioactive molecules (Bufadienolide (-12.30 kcal mol-1), Stigmasterol (-11.40 kcal mol-1), Isovitexin (-11.20 kcal mol-1), and Apigetrin (-11.20 kcal mol-1)) showed the top binding affinities with the PDE5 enzyme, much more powerful than the standard PDE5 inhibitor (-9.80 kcal mol-1). The four top binding bioactive molecules were further validated for a stable binding affinity with the PDE5 enzyme and conformation during the MD simulation period as compared to the apoprotein and standard PDE5 inhibitor complexes. Further, the four top binding bioactive molecules demonstrated significant drug-likeness characteristics with lower toxicity profiles. According to the findings, the four top binding molecules may be used as potent and safe PDE5 inhibitors and could potentially be used in the treatment of ED.


Asunto(s)
Afrodisíacos , Disfunción Eréctil , Mimosa , Afrodisíacos/uso terapéutico , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 5 , Disfunción Eréctil/tratamiento farmacológico , Humanos , Masculino , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Inhibidores de Fosfodiesterasa 5/química
13.
Int Microbiol ; 25(3): 629-638, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35554762

RESUMEN

Biofilm formation associated with quorum sensing (QS) is a community behaviour displayed by many gram-negative pathogenic bacteria that provide survival advantages in hostile conditions. The inhibitors of QS interrupt bacterial communication and coordinated cell signalling for community aggregation in the biofilm. Thymol, a natural monoterpenoid, was tested against QS in Chromobacterium violaceum. As the first step, the interaction of thymol with cviR protein was investigated using in silico approach followed by validation using detailed in vitro experiments. The QS and biofilm studies were performed using the wild type of strain C. violaceum ATCC 12,472 and a mini-Tn5 mutant CV026. The MIC of thymol was established by the broth micro-dilution method, and IC50 value for violacein inhibition was quantified spectrophotometrically by extracting the violacein from the treated cells. Inhibitory effect of thymol on the biofilm was quantified by the crystal violet staining method, and scanning electron microscopy (SEM) was employed for biofilm visualization. The expression of biofilm associated genes (hmsH, hmsR, pilB, and pilT) was evaluated by qRT-PCR analysis. The in silico molecular interactions of thymol with cviR exhibited a G-score of - 5.847 kcal/mol, binding with TYR-80 and SER-155 by Pi-Pi stacking and H-bond, respectively. The MIC of thymol was 160 µg/mL, and the IC50 for violacein inhibition was estimated to be 28 µg/mL. The thymol treatment significantly reduced the biofilm viability and biomass by > 80% along with disruption of the well-organized biofilm architecture. QS inhibitory activity of thymol resulted in the reduction of exopolysaccharide production, swarming motility, and downregulation of biofilm-associated hmsH, hmsR, pilB, and pilT genes. This data establishes the QS inhibitory role of thymol in the biofilm formation in C. violaceum.


Asunto(s)
Percepción de Quorum , Timol , Antibacterianos/química , Antibacterianos/farmacología , Biopelículas , Chromobacterium , Bacterias Gramnegativas , Extractos Vegetales/química , Timol/farmacología
14.
J Pharm Pharmacol ; 73(6): 749-757, 2021 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-33769483

RESUMEN

BACKGROUND: Taxane based conventional chemotherapy serves as the standard treatment regimen for triple-negative breast cancer (TNBC). However, the efficacy is plateaued due to toxicities, chemoresistance and metastasis. Hence, the development of new therapies that provide long-term cover is needed. Brusatol, a natural quassinoid, has been implicated to inhibit the migration and proliferation of metastatic cells in lung and liver carcinoma, but its efficacy in TNBC has not been explored. METHODS: The growth inhibitory activity on TNBC cells was measured using MTT assay and flow cytometry. Epithelial to mesenchymal transition (EMT) and apoptotic markers were quantified using western blotting. The caspases using Calorimetric assay. RESULTS: Brusatol along with paclitaxel showed an enhanced growth inhibitory activity and a combined synergistic effect. In addition, brusatol was also observed to inhibit the invasion, migratory potential of TNBC cells. Mechanistically, brusatol and its combination were observed to decrease the matrix metalloproteinase (MMP) and a modest increase in the reactive oxygen species (ROS) production. Furthermore, brusatol treatment activated both intrinsic and extrinsic pathways with morphological changes of apoptosis in TNBC cells. CONCLUSION: This is the first in vitro report demonstrating antineoplastic, anti-EMT and synergistic activity of brusatol and in combination with paclitaxel in TNBC cell. Further in-vivo studies are needed to substantiate the above findings.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Paclitaxel/farmacología , Cuassinas/farmacología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Humanos , Invasividad Neoplásica/prevención & control , Paclitaxel/administración & dosificación , Cuassinas/administración & dosificación , Especies Reactivas de Oxígeno/metabolismo , Neoplasias de la Mama Triple Negativas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...