Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Biol Chem ; 294(17): 6733-6750, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-30824542

RESUMEN

Notch signaling is reported to be deregulated in several malignancies, including breast, and the enzyme γ-secretase plays an important role in the activation and nuclear translocation of Notch intracellular domain (NICD). Hence, pharmacological inhibition of γ-secretase might lead to the subsequent inhibition of Notch signaling in cancer cells. In search of novel γ-secretase inhibitors (GSIs), we screened a series of triazole-based compounds for their potential to bind γ-secretase and observed that 3-(3'4',5'-trimethoxyphenyl)-5-(N-methyl-3'-indolyl)-1,2,4-triazole compound (also known as NMK-T-057) can bind to γ-secretase complex. Very interestingly, NMK-T-057 was found to inhibit proliferation, colony-forming ability, and motility in various breast cancer (BC) cells such as MDA-MB-231, MDA-MB-468, 4T1 (triple-negative cells), and MCF-7 (estrogen receptor (ER)/progesterone receptor (PR)-positive cell line) with negligible cytotoxicity against noncancerous cells (MCF-10A and peripheral blood mononuclear cells). Furthermore, significant induction of apoptosis and inhibition of epithelial-to-mesenchymal transition (EMT) and stemness were also observed in NMK-T-057-treated BC cells. The in silico study revealing the affinity of NMK-T-057 toward γ-secretase was further validated by a fluorescence-based γ-secretase activity assay, which confirmed inhibition of γ-secretase activity in NMK-T-057-treated BC cells. Interestingly, it was observed that NMK-T-057 induced significant autophagic responses in BC cells, which led to apoptosis. Moreover, NMK-T-057 was found to inhibit tumor progression in a 4T1-BALB/c mouse model. Hence, it may be concluded that NMK-T-057 could be a potential drug candidate against BC that can trigger autophagy-mediated cell death by inhibiting γ-secretase-mediated activation of Notch signaling.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Neoplasias de la Mama/patología , Receptores Notch/metabolismo , Transducción de Señal , Triazoles/farmacología , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Carcinogénesis/efectos de los fármacos , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos BALB C , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Cell Biochem ; 120(4): 5987-6003, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30390323

RESUMEN

Theaflavin (TF) and epigallocatechin-3-gallate (EGCG) both have been reported previously as microtubule depolymerizing agents that also have anticancer effects on various cancer cell lines and in animal models. Here, we have applied TF and EGCG in combination on HeLa cells to investigate if they can potentiate each other to improve their anticancer effect in lower doses and the underlying mechanism. We found that TF and EGCG acted synergistically, in lower doses, to inhibit the growth of HeLa cells. We found the combination of 50 µg/mL TF and 20 µg/mL EGCG to be the most effective combination with a combination index of 0.28. The same combination caused larger accumulation of cells in the G 2 /M phase of the cell cycle, potent mitochondrial membrane potential loss, and synergistic augmentation of apoptosis. We have shown that synergistic activity might be due to stronger microtubule depolymerization by simultaneous binding of TF and EGCG at different sites on tubulin: TF binds at vinblastine binding site on tubulin, and EGCG binds near colchicines binding site on tubulin. A detailed mechanistic analysis revealed that stronger microtubule depolymerization caused effective downregulation of PI3K/Akt signaling and potently induced mitochondrial apoptotic signals, which ultimately resulted in the apoptotic death of HeLa cells in a synergistic manner.


Asunto(s)
Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Biflavonoides/farmacología , Catequina/análogos & derivados , Microtúbulos/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Polimerizacion/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Antioxidantes/metabolismo , Biflavonoides/metabolismo , Sitios de Unión , Catequina/metabolismo , Catequina/farmacología , Proliferación Celular/efectos de los fármacos , Sinergismo Farmacológico , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Cabras , Células HeLa , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Tubulina (Proteína)/aislamiento & purificación , Tubulina (Proteína)/metabolismo
3.
Apoptosis ; 24(5-6): 434, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30923989

RESUMEN

The original version of this article unfortunately contained an error in acknowledgment text. The authors would like to include a statement: "Moumita Dasgupta is supported by Junior Research Fellowship from University Grant Commission, India." in acknowledgment section.

4.
Apoptosis ; 24(5-6): 414-433, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30767087

RESUMEN

Paclitaxel is one of the most commonly used drugs for the treatment of nonsmall cell lung cancer (NSCLC). However acquired resistance to paclitaxel, epithelial to mesenchymal transition and cancer stem cell formation are the major obstacles for successful chemotherapy with this drug. Some of the major reasons behind chemoresistance development include increased ability of the cancer cells to survive under stress conditions by autophagy, increased expression of drug efflux pumps, tubulin mutations etc. In this study we found that inhibition of autophagy with chloroquine prevented development of paclitaxel resistance in A549 cells with time and potentiated the effect of paclitaxel by increased accumulation of superoxide-producing damaged mitochondria, with elevated ROS generation, it also increased the apoptotic rate and sub G0/ G1 phase arrest with time in A549 cells treated with paclitaxel and attenuated the metastatic potential and cancer stem cell population of the paclitaxel-resistant cells by ROS mediated modulation of the Wnt/ß-catenin signaling pathway, thereby increasing paclitaxel sensitivity. ROS here played a crucial role in modulating Akt activity when autophagy process was hindered by chloroquine, excessive ROS accumulation in the cell inhibited Akt activity. In addition, chloroquine pre-treatment followed by taxol (10 nM) treatment did not show significant toxicity towards non-carcinomas WI38 cells (lung fibroblast cells). Thus autophagy inhibition by CQ pre-treatment can be used as a fruitful strategy to combat the phenomenon of paclitaxel resistance development as well as metastasis in lung cancer.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Autofagia/efectos de los fármacos , Cloroquina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Paclitaxel/farmacología , Especies Reactivas de Oxígeno/metabolismo , beta Catenina/metabolismo , Células A549 , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Vía de Señalización Wnt/efectos de los fármacos
5.
Nanomedicine ; 15(1): 47-57, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30213518

RESUMEN

Little is known about insulin's wound healing capability in normal as well as diabetic conditions. We here report specific interaction of silver nanoparticles (AgNPs) with insulin by making a ~2 nm thick coat around the AgNPs and its potent wound healing efficacy. Characterization of the interaction of human insulin with silver nanoparticles showed confirmed alteration of amide-I in insulin whereas amide-II and III remained unaltered. Further, nanoparticles protein interaction kinetics showed spontaneous interaction at physiological temperature with ΔG, ΔS, Ea and Ka values -7.48, 0.076, 3.84 kcal mol-1 and 6 × 105 s-1 respectively. Insulin loaded AgNPs (IAgNPs) showed significant improvement in healing activity in vitro (HEKa cells) and in vivo (Wister Rats) in comparison with the control in both normal and diabetic conditions. The underlying mechanism was attributed to a regulation of the balance between pro (IL-6, TNFα) and anti-inflammatory cytokines (IL-10) at the wound site to promote faster wound remodeling.


Asunto(s)
Citocinas/metabolismo , Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Insulina/farmacología , Nanopartículas del Metal/administración & dosificación , Cicatrización de Heridas , Animales , Movimiento Celular , Diabetes Mellitus Experimental/metabolismo , Composición de Medicamentos , Sistemas de Liberación de Medicamentos , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/química , Mediadores de Inflamación/metabolismo , Insulina/administración & dosificación , Insulina/química , Masculino , Nanopartículas del Metal/química , Ratas , Ratas Wistar , Plata/química
6.
Tumour Biol ; 39(2): 1010428317694314, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28240052

RESUMEN

Paclitaxel (Tx) is one of the first-line chemotherapeutic drugs used against lung cancer, but acquired resistance to this drug is a major challenge against successful chemotherapy. In this work, we have focused on the chronological changes of various cellular parameters and associated effect on Tx (10 nM) resistance development in A549 cell line. It was observed, at initial stage, the cell death percentage due to drug treatment had increased up to 20 days, and thereafter, it started declining and became completely resistant by 40 days. Expressions of ßIII tubulin and drug efflux pumps also increased over the period of resistance development. Changes in cellular autophagy and reactive oxygen species generation showed a biphasic pattern and increased gradually over the course of upto 20 days, thereafter declined gradually; however, their levels remained higher than untreated cells when resistance was acquired. Increase in extracellular acidification rates and oxygen consumption rates was found to be directly correlated with acquisition of resistance. The depolarisation of mitochondrial membrane potential was also biphasic; first, it increased with increase of cell death up to 20 days, thereafter, it gradually decreased to normal level along with resistance development. Increase in activity of catalase, glutathione peroxidase and glutathione content over these periods may attribute in bringing down the reactive oxygen species levels and normalisation of mitochondrial membrane potential in spite of comparatively higher reactive oxygen species production by the Tx-resistant cells.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/fisiología , Paclitaxel/farmacología , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Antineoplásicos Fitogénicos/farmacología , Autofagia , Caspasa 3/metabolismo , Ciclo Celular/fisiología , Resistencia a Antineoplásicos , Metabolismo Energético , Humanos , Neoplasias Pulmonares/patología , Microscopía Fluorescente
7.
Biochemistry ; 55(21): 3020-35, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27110637

RESUMEN

The biological significance of microtubules makes them a validated target of cancer therapy. In this study, we have utilized indole, an important pharmacological scaffold, to synthesize novel bis(indolyl)-hydrazide-hydrazone derivatives (NMK-BH compounds) and recognized NMK-BH3 as the most effective one in inhibiting A549 cell proliferation and assembly of tissue-purified tubulin. Cell viability experiments showed that NMK-BH3 inhibited proliferation of human lung adenocarcinoma (A549) cells, normal human lung fibroblasts (WI38) and peripheral blood mononuclear cells (PBMC) with IC50 values of ∼2, 48.5, and 62 µM, respectively. Thus, the relatively high cytotoxicity of NMK-BH3 toward lung carcinoma (A549) cells over normal lung fibroblasts (WI38) and PBMC confers a therapeutic advantage of reduced host toxicity. Flow cytometry, Western blot, and immunofluorescence studies in the A549 cell line revealed that NMK-BH3 induced G2/M arrest, mitochondrial depolarization, and apoptosis by depolymerizing the cellular interphase and spindle microtubules. Consistent with these observations, study in cell free system revealed that NMK-BH3 inhibited the microtubule assembly with an IC50 value of ∼7.5 µM. The tubulin-ligand interaction study using fluorescence spectroscopy indicated that NMK-BH3 exhibited strong and specific tubulin binding with a dissociation constant of ∼1.4 µM at a single site, very close to colchicine site, on ß-tubulin. Collectively, these findings explore the cytotoxic potential of NMK-BH3 by targeting the microtubules and inspire its development as a potential candidate for lung cancer chemotherapy.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Citotoxinas/farmacología , Hidrazonas/química , Indoles/química , Indoles/farmacología , Neoplasias Pulmonares/patología , Microtúbulos/efectos de los fármacos , Moduladores de Tubulina/farmacología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Antineoplásicos/química , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Citotoxinas/química , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Humanos , Hidrazonas/farmacología , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microscopía Fluorescente , Modelos Moleculares , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química
8.
Tumour Biol ; 37(8): 10653-64, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26867767

RESUMEN

Colchicine is a well-known and potent microtubule targeting agent, but the therapeutic value of colchicine against cancer is limited by its toxicity against normal cells. But, there is no report of its cytotoxic potential against lung cancer cell, at clinically permissible or lower concentrations, minimally toxic to non-cancerous cells. Hence, in the present study, we investigated the possible mechanism by which the efficacy of colchicine against lung cancer cells at less toxic dose could be enhanced. Colchicine at clinically admissible concentration of 2.5 nM had no cytotoxic effect and caused no G2/M arrest in A549 cells. However, at this concentration, colchicine strongly hindered the reformation of cold depolymerised interphase and spindle microtubule. Colchicine induced senescence and reactive oxygen species mediated autophagy in A549 cells at this concentration. Autophagy inhibitor 3-methyladenine (3-MA) sensitised the cytotoxicity of colchicine in A549 cells by switching senescence to apoptotic death, and this combination had reduced cytotoxicity to normal lung fibroblast cells (WI38). Together, these findings indicated the possible use of colchicine at clinically relevant dose along with autophagy inhibitor in cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Autofagia/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Colchicina/farmacología , Neoplasias Pulmonares/patología , Células A549 , Adenina/análogos & derivados , Adenina/farmacología , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Línea Celular , Colchicina/administración & dosificación , Colchicina/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Fibroblastos , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Huso Acromático/efectos de los fármacos , Moduladores de Tubulina/administración & dosificación , Moduladores de Tubulina/antagonistas & inhibidores , Moduladores de Tubulina/farmacología
9.
Cancer Lett ; 585: 216662, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38309614

RESUMEN

Maintaining cellular homeostasis relies on the interplay between apoptosis and autophagy, and disruption in either of these processes can contribute to the development of cancer. Autophagy can hinder the apoptotic process, and when autophagy is inhibited in such instances, it can enhance the rate of apoptosis. However, evidence suggests that excessive autophagy can also lead to apoptotic cell death. Also, excess autophagy can cause excessive digestion of cellular organelles, causing autophagic cell death. Targeting autophagy in non-small cell lung cancer (NSCLC), the most common form of lung cancer, can be very tricky due to the dual nature of autophagy. According to genetic analysis, various mutations in p53 and EGFR, G:C to A:T transversions seem responsible for the development of lung cancer in smokers and non-smokers. These events trigger cytoprotective autophagy or induce apoptotic cell death through different but interconnected signalling pathways. Lung cancer being the leading cause of death worldwide, calls for more attention to disease prognosis and new therapeutics in the market. However, molecules responsible for autophagy to apoptosis transition are yet to be studied elaborately. Also, the role of effector caspases during this shift needs to be elucidated in future. To comprehend how therapeutics operate through the modulation of autophagy and apoptosis and to target such pathways, it is crucial to emphasize these intricate connections. Many therapeutics discussed in this review targeting both apoptosis and autophagy have shown promising results in vitro and in vivo, however, few have crossed the hurdles of clinical trial. Nevertheless, the quest for safer and better efficacious agents is still alive, with the sole aim to develop novel cancer chemotherapeutic(s).


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Apoptosis/genética , Transducción de Señal , Autofagia/genética , Línea Celular Tumoral
10.
Curr Drug Deliv ; 20(10): 1441-1464, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36200202

RESUMEN

COVID-19 pandemic is the biggest global crisis. The frequent mutations in coronavirus to generate new mutants are of major concern. The pathophysiology of SARS-CoV-2 infection has been well studied to find suitable molecular targets and candidate drugs for effective treatment. FDArecommended etiotropic therapies are currently followed along with mass vaccination. The drug delivery system and the route of administration have a great role in enhancing the efficacy of therapeutic agents and vaccines. Since COVID-19 primarily infects the lungs in the affected individuals, pulmonary administration may be the best possible route for the treatment of COVID-19. Liposomes, solid lipid nanoparticles, polymeric nanoparticles, porous microsphere, dendrimers, and nanoparticles encapsulated microparticles are the most suitable drug delivery systems for targeted drug delivery. The solubility, permeability, chemical stability, and biodegradability of drug molecules are the key factors for the right selection of suitable nanocarriers. The application of nanotechnology has been instrumental in the successful development of mRNA, DNA and subunit vaccines, as well as the delivery of COVID-19 therapeutic agents.


Asunto(s)
COVID-19 , Humanos , Pandemias/prevención & control , SARS-CoV-2 , Sistemas de Liberación de Medicamentos , Vacunas contra la COVID-19
11.
Toxicol In Vitro ; 93: 105703, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37751786

RESUMEN

OBJECTIVE: Deuterium oxide (D2O) or heavy water is known to have diverse biological activities and have a few therapeutic applications due to its limited toxicity to human subjects. In the present study, we investigated the mechanism of D2O-induced cytotoxicity in non-small cell lung cancer A549 cells. RESULTS: We found that D2O-treatment resulted in cytotoxicity, cell cycle arrest, and apoptosis in A549 cells in a dose-dependent fashion. In contrast, limited cytotoxicity was observed in lung fibroblasts WI38 cells. Moreover, D2O-treatment resulted in the disruption of the cellular microtubule network, accompanied by the generation of ROS. On further investigation, we observed that the intracellular ROS triggered autophagic responses in D2O-treated cells, leading to apoptosis by inhibiting the oncogenic PI3K/ Akt/ mTOR signaling. D2O-treatment was also found to enhance the efficacy of paclitaxel in A549 cells. SIGNIFICANCE: D2O induces autophagy-dependent apoptosis in A549 cells via ROS generation upon microtubule depolymerization and inhibition of PI3K/ Akt/ mTOR signaling. It augments the efficacy of other microtubule-targeting anticancer drug taxol, which indicates the potential therapeutic importance of D2O as an anticancer agent either alone or in combination with other chemotherapeutic drugs.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Células A549 , Óxido de Deuterio/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Apoptosis , Autofagia , Microtúbulos , Fosfatidilinositol 3-Quinasas/metabolismo
12.
Sci Rep ; 12(1): 6241, 2022 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-35422113

RESUMEN

Recently published clinical data from COVID-19 patients indicated that statin therapy is associated with a better clinical outcome and a significant reduction in the risk of mortality. In this study by computational analysis, we have aimed to predict the possible mechanism of the statin group of drugs by which they can inhibit SARS-CoV-2 pathogenesis. Blind docking of the critical structural and functional proteins of SARS-CoV-2 like RNA-dependent RNA polymerase, M-protease of 3-CL-Pro, Helicase, and the Spike proteins ( wild type and mutants from different VOCs) were performed using the Schrodinger docking tool. We observed that fluvastatin and pitavastatin showed fair, binding affinities to RNA polymerase and 3-CL-Pro, whereas fluvastatin showed the strongest binding affinity to the helicase. Fluvastatin also showed the highest affinity for the SpikeDelta and a fair docking score for other spike variants. Additionally, molecular dynamics simulation confirmed the formation of a stable drug-protein complex between Fluvastatin and target proteins. Thus our study shows that of all the statins, fluvastatin can bind to multiple target proteins of SARS-CoV-2, including the spike-mutant proteins. This property might contribute to the potent antiviral efficacy of this drug.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Antivirales/uso terapéutico , Fluvastatina/farmacología , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , SARS-CoV-2
13.
Apoptosis ; 16(9): 924-39, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21667044

RESUMEN

Naphthazarin (DHNQ, 5,8-dihydroxy-l,4-naphthoquinone) is a naturally available 1,4-naphthoquinone derivatives. In this study, we focused on elucidating the cytotoxic mechanism of naphthazarin in A549 non-small cell lung carcinoma cells. Naphthazarin reduced the A549 cell viability considerably with an IC(50) of 16.4 ± 1.6 µM. Naphthazarin induced cell death in a dose- and time-dependent manner by activating apoptosis and autophagy pathways. Specifically, we found naphthazarin inhibited the PI3K/Akt cell survival signalling pathway, measured by p53 and caspase-3 activation, and PARP cleavage. It also resulted in an increase in the ratio of Bax/Bcl2 protein levels, indicating activation of the mitochondrial apoptotic pathway. Similarly naphthazarin triggered LC3II expression and induced autophagic flux in A549 cells. We demonstrated further that naphthazarin is a microtubule inhibitor in cell-free system and in A549 cells. Naphthazarin treatment depolymerized interphase microtubules and disorganised spindle microtubules and the majority of cells arrested at the G(2)/M transition. Together, these data suggest that naphthazarin, a microtubule depolymerizer which activates dual cell death machineries, could be a potential novel chemotherapeutic agent.


Asunto(s)
Apoptosis , Autofagia , Microtúbulos/efectos de los fármacos , Naftoquinonas/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Caspasa 3/metabolismo , Ciclo Celular , Puntos de Control del Ciclo Celular , Línea Celular Tumoral/efectos de los fármacos , Supervivencia Celular , Sistema Libre de Células/metabolismo , Relación Dosis-Respuesta a Droga , Citometría de Flujo , Humanos , Inmunoprecipitación , Concentración 50 Inhibidora , Microscopía Confocal , Microtúbulos/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Índice Mitótico , Estructura Molecular , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Tubulina (Proteína)/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
14.
Biochim Biophys Acta Gen Subj ; 1865(8): 129931, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34023444

RESUMEN

BACKGROUND: Increasing antibiotic-resistance in bacterial strains has boosted the need to find new targets for drug delivery. FtsA, a major bacterial divisome protein can be a potent novel drug-target. METHODS AND RESULTS: This study finds, morin (3,5,7,2',4'-pentahydroxyflavone), a bio-available flavonoid, had anti-bacterial activities against Vibrio cholerae, IC50 (50 µM) and MIC (150 µM). Morin (2 mM) kills ~20% of human lung fibroblast (WI38) and human intestinal epithelial (HIEC-6) cells in 24 h in-vitro. Fluorescence studies showed morin binds to VcFtsA (FtsA of V. cholerae) with a Kd of 4.68 ± 0.4 µM, inhibiting the protein's polymerization by 72 ± 7% at 25 µM concentration. Morin also affected VcFtsA's ATPase activity, recording ~80% reduction at 20 µM concentration. The in-silico binding study indicated binding sites of morin and ATP on VcFtsA had overlapping amino acids. Mant-ATP, a fluorescent ATP-derivative, showed increased fluorescence on binding to VcFtsA in absence of morin, but in its presence, Mant-ATP fluorescence decreased. VcFtsA-S40A mutant protein did not bind to morin. CONCLUSIONS: VcFtsA-morin interaction inhibits the polymerization of the protein by affecting its ATPase activity. The destabilized VcFtsA assembly in-turn affected the cell division in V. cholerae, yielding an elongated morphology. GENERAL SIGNIFICANCE: Collectively, these findings explore the anti-bacterial effect of morin on V. cholerae cells targeting VcFtsA, encouraging it to become a potent anti-bacterial agent. Low cytotoxicity of morin against human cells (host) is therapeutically advantageous. This study will also help in synthesizing novel derivatives that can target VcFtsA more efficiently.


Asunto(s)
Adenosina Trifosfato/metabolismo , Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Flavonoides/farmacología , Regulación Bacteriana de la Expresión Génica , Vibrio cholerae/efectos de los fármacos , Proteínas Bacterianas/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/microbiología , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/microbiología , Unión Proteica , Vibrio cholerae/metabolismo
15.
Biochemistry ; 49(8): 1702-12, 2010 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-20085293

RESUMEN

Genistein (4',5,7-trihydroxyisoflavone), an isoflavone, is a major constituent of soyfoods. It has potential antiproliferative activity against several tumor types. We have examined the effect of genistein on cellular microtubules as well as its binding with purified tubulin in vitro. Cell viability experiments using human non-small lung epithelium carcinoma cells (A549) indicated that the IC(50) value for genistein is 72 microM. Flow cytometry experiments demonstrated that genistein arrested cell cycle progression at the G(2)/M phase, but mitotic index data showed that genistein did not arrest cell cycle progression at mitosis. Immunofluorescence studies using an anti-alpha-tubulin antibody demonstrated a significant depolymerization of the interphase microtubules in a dose-dependent manner, and this was confirmed by the Western blot experiment using genistein-treated A549 cells. In vitro polymerization of purified tubulin into microtubules was inhibited by genistein with an IC(50) value of 87 microM. Genistein binding to tubulin quenched protein tryptophan fluorescence in a time- and concentration-dependent manner. Binding of genistein to tubulin was slow, taking approximately 45 min for equilibration at 37 degrees C. The association rate constant was 104.64 +/- 20.63 M(-1) s(-1) at 37 degrees C. The stoichiometry of genistein binding to tubulin was nearly 1:1 (molar ratio) with a dissociation constant of 15 microM at 37 degrees C. It was interesting to note that genistein did not recognize either the colchicine site or the vinblastine binding site of tubulin. Surprisingly, genistein inhibited ANS binding and competed for its binding site of tubulin with a K(i) of 20 microM as determined from a modified Dixon plot. Hence, we conclude that one of the mechanisms of antiproliferative activity of genistein is depolymerization of microtubules through binding of tubulin.


Asunto(s)
Ciclo Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Fase G2/efectos de los fármacos , Genisteína/farmacología , Interfase/efectos de los fármacos , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Western Blotting , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Genisteína/química , Humanos , Microscopía Electrónica de Transmisión , Microtúbulos/química , Microtúbulos/ultraestructura , Estructura Molecular , Unión Proteica , Tubulina (Proteína)/química , Tubulina (Proteína)/ultraestructura
16.
Chem Res Toxicol ; 23(6): 1054-66, 2010 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-20499891

RESUMEN

Parabenzoquinone (1,4-benzoquinone) (PBQ) is a bioactve quinone present in cigarette smoke and diesel smoke, which causes severe genotoxic effects both in vitro and in vivo. In the previous study, we showed that the microtubules are one of the major targets of cigarette smoke-induced damage of lung epithelium cells. In the present study, we have investigated the effect of PBQ on cellular microtubules using human type II lung epithelial cells (A549) and also on purified tubulin. Cell viability experiments using A549 cells indicated a very low IC(50) value (approximately 7.5 microM) for PBQ. PBQ inhibited cell cycle progression and induced apoptosis of A549 cells. PBQ also induced the contraction and shrinkage of the A549 cells in a time- and concentration-dependent manner, which is proved to be a direct effect of the damage of the microtubule cytoskeleton network, and that was demonstrated by a immunofluorescence study. PBQ also inhibited the assembly of tubulin in lung cells and a in cell free system (IC(50) approximately 5 microM). Treatment with PBQ resulted in the degradation of tubulin in lung cells without affecting the actin network, and this was confirmed by a Western blot experiment. Upregulation of pro-apoptotic proteins such as p53 and Bax and downregulation of antiapoptotic protein Bcl-2 were observed in PBQ-treated A549 cells. Simultaneously, loss of mitochondrial membrane potential and activation of caspase-3 were also observed in the PBQ treated lung epithelium cells. Fluorescence and circular dichroism studies demonstrated that the denaturation of tubulin in a cell free system was caused by PBQ. However, in the presence of N-acetyl cysteine (NAC), damage of the microtubule network in A549 cells by PBQ was prevented, which led to a significant increase in the viability of A549 cells. These results suggest that microtubule damage is one of the key mechanisms of PBQ induced cytotoxity in lung cells.


Asunto(s)
Benzoquinonas/efectos adversos , Caspasa 3/metabolismo , Células Epiteliales/efectos de los fármacos , Pulmón/citología , Microtúbulos/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular , Activación Enzimática/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Humanos
17.
J Basic Microbiol ; 50(6): 519-26, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20806243

RESUMEN

Of the three cold shock proteins expressed by Staphylococcus aureus, CspC is induced poorly by cold but strongly by various antibiotics and toxic chemicals. Using a purified CspC, here we demonstrate that it exists as a monomer in solution, possesses primarily ß-sheets, and bears substantial structural similarity with other bacterial Csps. Aggregation of CspC was initiated rapidly at temperatures above 40 °C, whereas, the Gibbs free energy of stabilization of CspC at 0 M GdmCl was estimated to be +1.6 kcal mol(-1), indicating a less stable protein. Surprisingly, CspC showed stable binding with ssDNA carrying a stretch of more than three thymine bases and binding with such ssDNA had not only stabilized CspC against proteolytic degradation but also quenched the fluorescence intensity from its exposed Trp residue. Analysis of quenching data indicates that each CspC molecule binds with ∼5 contiguous thymine bases of the above ssDNA and binding is cooperative in nature.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Staphylococcus aureus/metabolismo , Staphylococcus aureus/efectos de la radiación , Antibacterianos/toxicidad , Proteínas Bacterianas/aislamiento & purificación , Dicroismo Circular , Frío , ADN de Cadena Simple/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Proteínas de Choque Térmico/aislamiento & purificación , Modelos Moleculares , Unión Proteica , Conformación Proteica , Desnaturalización Proteica , Estabilidad Proteica , Timina/metabolismo
18.
Int J Biol Macromol ; 142: 18-32, 2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31790740

RESUMEN

Proper interaction between the divisome proteins FtsA and FtsZ is important for the bacterial cell division which is not well characterized till date. In this study, the objective was to understand the mechanism of FtsA-FtsZ interaction using full-length recombinant proteins. We cloned, over-expressed, purified and subsequently characterized FtsA of Vibrio cholerae (VcFtsA). We found that VcFtsA polymerization assembly was dependent on Ca2+ ions, which is unique among FtsA proteins reported until now. VcFtsA also showed ATPase activity and its assembly was ATP dependent. Binding parameters of the interaction between the two full-length proteins, VcFtsA, and VcFtsZ determined by fluorescence spectrophotometry yielded a Kd value of around 38 µM. The Kd value of the interaction was 3 µM when VcFtsA was in ATP bound state. We found that VcFtsZ after interacting with VcFtsA causes a change of secondary structure in the later one leading to loss of its ability to hydrolyze ATP, subsequently halting the VcFtsA polymerization. On the other hand, a double-mutant of VcFtsA (VcFtsA-D242E,R300E), that does not bind to VcFtsZ, polymerized in the presence of VcFtsZ. Though FtsA proteins among different organisms show 70-80% homology in their sequences, assembly of VcFtsA showed a difference in its regulatory processes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Proteínas del Citoesqueleto/química , Polimerizacion , Vibrio cholerae/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Calcio , Clonación Molecular , Proteínas del Citoesqueleto/metabolismo , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Estructura Secundaria de Proteína , Proteínas Recombinantes , Análisis de Secuencia de Proteína
19.
Mater Sci Eng C Mater Biol Appl ; 106: 110160, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31753371

RESUMEN

Cu-Ag-ZnO nanocomposite (NC) has been successfully synthesized by mechanical alloying the Cu, Zn and Ag powder mixture under Ar atmosphere within 4 h of milling. The nanocomposite is then conjugated with the antifungal drug fluconazole by adding 5 wt% powdered drug to the NC and mechanical alloying the total powder mixture for one more hour. The Rietveld refinement of XRD data and FTIR spectrum analyses reveal the detailed structural and microstructural characterizations of the nanocomposite-drug conjugate (NC-DC). Presence of Cu, Ag, ZnO and drug in the 5 h milled powder are confirmed by analyzing TEM images and FESEM-EDS spectrum. Results obtained from FESEM and TEM images reveal the measure of particle size of the nanocomposite-drug conjugate and it agrees well with the crystallite size obtained from the Rietveld refinement. A significant antifungal activity of NC-DC against Candida sp. fungi has been revealed using disk agar diffusion method. Minimum inhibitory concentration (MIC) test confirms that NC-DC with only 5 wt% fluconazole produces similar antifungal activity of the pure (100 wt%) and conventional fluconazole. Thus, the conjugation of conventional drug to a nanocomposite results in enhancement of drug efficiency by a factor 20 folds. This is very important, particularly, for those antibiotics which are very effective in controlling several epidemic diseases but show intense side effects when used at higher dose and/or for a longer duration.


Asunto(s)
Antifúngicos/química , Cobre/química , Fluconazol/química , Nanocompuestos/química , Plata/química , Óxido de Zinc/química , Antifúngicos/farmacología , Candida/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Fluconazol/farmacología , Humanos , Pruebas de Sensibilidad Microbiana , Tamaño de la Partícula
20.
Biochim Biophys Acta Mol Cell Res ; 1867(10): 118762, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32502617

RESUMEN

BACKGROUND: Microtubules, the key components of the eukaryotic cytoskeleton and mitotic spindle, are one of the most sought-after targets for cancer chemotherapy, especially due to their indispensible role in mitosis. Cervical cancer is a prevalent malignancy among women of developing countries including India. In spite of the remarkable therapeutic advancement, the non-specificity of chemotherapeutic drugs adversely affect the patients' survival and well-being, thus, necessitating the quest for novel indole-based anti-microtubule agent against cervical cancer, with high degree of potency and selectivity. METHODS: For in vitro studies, we used MTT assay, confocal microscopy, fluorescence microscopy, flow cytometry and Western blot analysis. Study in cell free system was accomplished by spectrophotometry, fluorescence spectroscopy and TEM and computational analysis was done by AutodockTools 1.5.6. RESULTS: NMK-BH2 exhibited significant and selective anti-proliferative activity against cervical cancer HeLa cells (IC50 = 1.5 µM) over normal cells. It perturbed the cytoskeletal and spindle microtubules of HeLa cells leading to mitotic block and cell death by apoptosis and autophagy. Furthermore, NMK-BH2 targeted the tubulin-microtubule system through fast and strong binding to the αß-tubulin heterodimers at colchicine-site. CONCLUSION: This study identifies and characterises NMK-BH2 as a novel anti-microtubule agent and provides insights into its key anti-cancer mechanism through two different cell death pathways: apoptosis and autophagy, which are mutually independent. GENERAL SIGNIFICANCE: It navigates the potential of the novel bis (indolyl)-hydrazide-hydrazone, NMK-BH2, to serve as lead for development of new generation microtubule-disrupting chemotherapeutic with improved efficacy and remarkable selectivity towards better cure of cervical cancer.


Asunto(s)
Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Colchicina/metabolismo , Hidrazonas/farmacología , Microtúbulos/metabolismo , Polimerizacion , Tubulina (Proteína)/metabolismo , Neoplasias del Cuello Uterino/patología , Animales , Sitios de Unión , Biomarcadores de Tumor/metabolismo , Proliferación Celular/efectos de los fármacos , Sistema Libre de Células , Simulación por Computador , Femenino , Cabras , Células HeLa , Humanos , Hidrazonas/química , Cinética , Ligandos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Microtúbulos/ultraestructura , Mitosis/efectos de los fármacos , Espectrometría de Fluorescencia , Huso Acromático/efectos de los fármacos , Huso Acromático/metabolismo , Tubulina (Proteína)/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA