Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Mol Biol Rep ; 51(1): 523, 2024 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-38630183

RESUMEN

BACKGROUND: In recent decades, phytotherapy has remained as a key therapeutic option for the treatment of various cancers. Evodiamine, an excellent phytocompound from Evodia fructus, exerts anticancer activity in several cancers by modulating drug resistance. However, the role of evodiamine in cisplatin-resistant NSCLC cells is not clear till now. Therefore, we have used evodiamine as a chemosensitizer to overcome cisplatin resistance in NSCLC. METHODS: Here, we looked into SOX9 expression and how it affects the cisplatin sensitivity of cisplatin-resistant NSCLC cells. MTT and clonogenic assays were performed to check the cell proliferation. AO/EtBr and DAPI staining, ROS measurement assay, transfection, Western blot analysis, RT-PCR, Scratch & invasion, and comet assay were done to check the role of evodiamine in cisplatin-resistant NSCLC cells. RESULTS: SOX9 levels were observed to be higher in cisplatin-resistant A549 (A549CR) and NCI-H522 (NCI-H522CR) compared to parental A549 and NCI-H522. It was found that SOX9 promotes cisplatin resistance by regulating ß-catenin. Depletion of SOX9 restores cisplatin sensitivity by decreasing cell proliferation and cell migration and inducing apoptosis in A549CR and NCI-H522CR. After evodiamine treatment, it was revealed that evodiamine increases cisplatin-induced cytotoxicity in A549CR and NCI-H522CR cells through increasing intracellular ROS generation. The combination of both drugs also significantly inhibited cell migration by inhibiting epithelial to mesenchymal transition (EMT). Mechanistic investigation revealed that evodiamine resensitizes cisplatin-resistant cells toward cisplatin by decreasing the expression of SOX9 and ß-catenin. CONCLUSION: The combination of evodiamine and cisplatin may be a novel strategy for combating cisplatin resistance in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Quinazolinas , Humanos , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Cisplatino/farmacología , beta Catenina , Transición Epitelial-Mesenquimal , Especies Reactivas de Oxígeno , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Muerte Celular , Factor de Transcripción SOX9/genética
2.
Cell Biol Toxicol ; 39(1): 1-31, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36138312

RESUMEN

Evodiamine is a major alkaloid component found in the fruit of Evodia rutaecarpa. It shows the anti-proliferative potential against a wide range of cancers by suppressing cell growth, invasion, and metastasis and inducing apoptosis both in vitro and in vivo. Evodiamine shows its anticancer potential by modulating aberrant signaling pathways. Additionally, the review focuses on several therapeutic implications of evodiamine, such as epigenetic modification, cancer stem cells, and epithelial to mesenchymal transition. Moreover, combinatory drug therapeutics along with evodiamine enhances the anticancer efficacy of chemotherapeutic drugs in various cancers by overcoming the chemo resistance and radio resistance shown by cancer cells. It has been widely used in preclinical trials in animal models, exhibiting very negligible side effects against normal cells and effective against cancer cells. The pharmacokinetic and pharmacodynamics-based collaborations of evodiamine are also included. Due to its poor bioavailability, synthetic analogs of evodiamine and its nano capsule have been formulated to enhance its bioavailability and reduce toxicity. In addition, this review summarizes the ongoing research on the mechanisms behind the antitumor potential of evodiamine, which proposes an exciting future for such interests in cancer biology.


Asunto(s)
Alcaloides , Antineoplásicos , Neoplasias , Animales , Transición Epitelial-Mesenquimal , Línea Celular Tumoral , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Alcaloides/farmacología , Extractos Vegetales/farmacología , Neoplasias/tratamiento farmacológico
3.
J Cell Biochem ; 123(9): 1454-1466, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35788981

RESUMEN

Evodiamine (EVO), a natural dietary alkaloid extracted from the roots of the Evodia rutaecarpa, has shown anticancer activities. Here, we have investigated EVO's role in inhibiting cell proliferation and migration in A549 and NCI-H522 lung cancer cells. EVO decreased the cell viability in A549 and NCI-H522 cells in a dose- and time-dependent manner. It also induced apoptosis by downregulating the expression of antiapoptotic Bcl-2 and upregulating the expression of cleaved caspase-3 and PARP. In addition, the treatment of EVO elevated the level of reactive oxygen species (ROS) generation inside the cells to induce the cell death pathways. In contrast, the pretreatment of ROS scavenger, N-acetyl cysteine, reverses the effect of EVO and attenuates cell death. Moreover, excess ROS generation in response to EVO resulted in the depletion of mitochondrial membrane potential. Furthermore, it induced DNA damage and arrested the cell cycle at the G2/M phase in A549 and NCI-H522 cells. Our study also investigated that EVO significantly suppressed tumorigenicity by inhibiting colony formation and tumorsphere formation. However, the treatment of EVO downregulated the cancer stem cell markers CD44 and CD133 in non-small-cell lung cancer. The inhibitory effect of EVO on cell invasion was mediated by altering the expression of E-cadherin, ZO-1, N-cadherin, and Vimentin. Additionally, we have revealed that EVO treatment showed downregulation of SOX9, an upstream component of ß-catenin. Lastly, we have demonstrated that EVO targets the SOX9-ß-catenin axis by reducing SOX9 and ß-catenin expression. These findings suggested that EVO could be a promising agent for treating human lung cancer.


Asunto(s)
Alcaloides , Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Alcaloides/farmacología , Apoptosis , Cadherinas , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Caspasa 3/metabolismo , Línea Celular Tumoral , Proliferación Celular , Cisteína , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Quinazolinas , Especies Reactivas de Oxígeno , Factor de Transcripción SOX9 , Vimentina , beta Catenina/metabolismo
4.
Bioconjug Chem ; 33(11): 2113-2120, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36264777

RESUMEN

Selective fluorescence imaging of actin protein hugely depends on the fluorescently labeled actin-binding domain (ABD). Thus, it is always a challenging task to image the actin protein (in vivo or in vitro) directly with an ABD-free system. To overcome the limitations of actin imaging without an ABD, we have designed a facile and cost-effective red fluorescent coumarin dye 7-hydroxy-4-methyl-8-(4-(2-oxo-2H-chromen-3-yl)thiazol-2-ylimino)methyl-2H-chromen-2-one (CTC) for actin binding. The selective binding of the dye was investigated using the gut and eye of the model organism Drosophila melanogaster and C2C12 and SCC-9 cell lines. Our result suggests two major advantages of CTC over the dyes presently used for imaging actin proteins. First, the dye can bind to actin efficiently without any secondary intermediate. Second, it is much more stable at room temperature and exhibits excellent photostability. To the best of our knowledge, this is the first fluorescent dye that can bind to the actin protein without employing any secondary intermediate/actin-binding domain. These findings could pave the way for many biologists and physicists to successfully employ the CTC dye for imaging and tracking actin proteins by fluorescence microscopy in various in vivo and in vitro systems.


Asunto(s)
Actinas , Colorantes Fluorescentes , Animales , Actinas/metabolismo , Colorantes Fluorescentes/química , Drosophila melanogaster , Cumarinas/química , Línea Celular
5.
Mol Biol Rep ; 47(6): 4155-4168, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32444975

RESUMEN

Plumbagin (PL) is a natural naphthoquinone compound, isolated from Plumbago zeylanica that has cytotoxic and antimigratory potential in many cancer. However, the cytotoxic mechanism of plumbagin in drug resistant lung cancer is poorly understood. To reveal the mechanism, we studied the anticancer effect of plumbagin in both gefitinib-sensitive and resistant A549 lung cancer cells. The anticancer potential of PL was demonstrated by MTT assay and the result suggested that PL showed cytotoxicity in both gefitinib-sensitive (A549) and gefitinib-resistant (A549GR) lung cancer cells. IC50 values of PL in A549 and A549GR were 3.2 µM and 4.5 µM, respectively. Morphological changes were also observed after treatment with PL. Furthermore, PL decreased cell survival by inhibiting colony formation ability, and inhibited cell migration at very low concentrations. From Annexin V-FITC/PI, AO/EtBr, and DAPI staining, we found that increasing concentration of PL leads to increase in apoptosis of lung cancer cells. Furthermore, western blotting results suggested that Bax and Caspase 3 levels were upregulated after PL treatment. In addition, treatment of PL caused DNA damage in a dose-dependent manner. PL arrested the cell cycle at S-G2/M phase, and enhanced reactive oxygen species (ROS) generation. Excess ROS generated by PL disrupted mitochondrial membrane resulted in depletion of mitochondrial membrane potential (MMP). These results conclude that PL decreases lung cancer cell viability by arresting cells at S-G2/M phase, and induces apoptosis by activation of mitochondrial-mediated apoptotic pathway through excess ROS generation. Overall findings suggest that plumbagin shows cytotoxic and therapeutic potential against both A549 and A549GR cell lines.


Asunto(s)
Células A549/efectos de los fármacos , Naftoquinonas/metabolismo , Naftoquinonas/farmacología , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Gefitinib/farmacología , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
Pharmacol Res ; 147: 104331, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31254665

RESUMEN

Lung cancer is the most common cancer-related death worldwide. Natural compounds have shown high biological and pharmaceutical relevance as anticancer agents. Retinoids are natural derivatives of vitamin A having many regulatory functions in the human body, including vision, cellular proliferation and differentiation, and activation of tumour suppressor genes. Retinoic acid (RA) is a highly active retinoid isoform with promising anti-lung cancer activity. The abnormal expression of retinoid receptors is associated with loss of anticancer activities and acquired resistance to RA in lung cancer. The preclinical promise has not translated to the general clinical utility of retinoids for lung cancer patients, especially those with a history of smoking. Newer retinoid nano-formulations and the combinatorial use of retinoids has been associated with lower toxicity and more favorably efficacy in both the preclinical and clinical settings. Here, we highlight epidemiological and clinical therapeutic studies involving retinoids and lung cancer. We also discuss the biological actions of retinoids in lung cancer, which include effects on cancer stem cell differentiation, angiogenesis, metastasis, and proliferative. We suggest that the use of retinoids in combination with conventional and targeted anticancer agents will broaden the utility of these potent anticancer compounds in the lung cancer clinic.


Asunto(s)
Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Retinoides/uso terapéutico , Animales , Humanos
7.
Mol Biol Rep ; 46(5): 5645-5659, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31280421

RESUMEN

Drug resistance is a major setback for advanced therapeutics in multiple cancers. The increasing prevalence of this resistance is a growing concern and bitter headache for the researchers since a decade. Hence, it is essential to revalidate the existing strategies available for cancer treatment and to look after a novel therapeutic approach for target based killing of cancer cells at the genetic level. This review outlines the different mechanisms enabling resistance including drug efflux, drug target alternation, alternative splicing, the release of the extracellular vesicle, tumor heterogeneity, epithelial-mesenchymal transition, tumor microenvironment, the secondary mutation in the receptor, epigenetic alternation, heterodimerization of receptors, amplification of target and amplification of components rather than the target. Furthermore, existing evidence and the role of various signaling pathways like EGFR, Ras, PI3K/Akt, Wnt, Notch, TGF-ß, Integrin-ECM signaling in drug resistance are explained. Lastly, the prevention of this resistance by a contemporary therapeutic strategy, i.e., a combination of specific signaling pathway inhibitors and the cocktail of a cancer drug is summarized showing the new treatment strategies.


Asunto(s)
Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/fisiología , Neoplasias/tratamiento farmacológico , Antineoplásicos/farmacología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/efectos de los fármacos , Genes erbB-1/fisiología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Microambiente Tumoral/efectos de los fármacos , Vía de Señalización Wnt/fisiología
8.
Biotechnol J ; 19(2): e2300370, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38375578

RESUMEN

Exosomes have been the hidden treasure of the cell in terms of cellular interactions, transportation and therapy. The native exosomes (NEx) secreted by the parent cells hold promising aspects in cancer diagnosis and therapy. NEx has low immunogenicity, high biocompatibility, low toxicity and high stability which enables them to be an ideal prognostic biomarker in cancer diagnosis. However, due to heterogeneity, NEx lacks specificity and accuracy to be used as therapeutic drug delivery vehicle in cancer therapy. Transforming these NEx with their innate structure and multiple receptors to engineered exosomes (EEx) can provide better opportunities in the field of cancer theranostics. The surface of the NEx exhibits numeric receptors which can be modified to pave the direction of its therapeutic drug delivery in cancer therapy. Through surface membrane, EEx can be modified with increased drug loading potentiality and higher target specificity to act as a therapeutic nanocarrier for drug delivery. This review provides insights into promising aspects of NEx as a prognostic biomarker and drug delivery tool along with its need for the transformation to EEx in cancer theranostics. We have also highlighted different methods associated with NEx transformations, their nano-bio interaction with recipient cells and major challenges of EEx for clinical application in cancer theranostics.


Asunto(s)
Exosomas , Neoplasias , Humanos , Exosomas/química , Medicina de Precisión , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Biomarcadores/metabolismo
9.
Arch Oral Biol ; 151: 105697, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37079976

RESUMEN

OBJECTIVE: Oral cancer is the sixteenth most prevalent cancer in the world and the third-most in India. Despite of several treatment modalities, the cure rate of oral cancer is still low due to drug resistance mechanisms, which are caused by many reasons. It is necessary to improve the existing treatment strategies and discover neoteric therapy to kill cancer cells, which will give oral cancer's cure rate more success. So this review aims to delineate the molecular mechanisms behind cisplatin resistance, specifically the role of the tumor microenvironment, extracellular vesicles, and altered signaling pathways and its overcoming strategies in oral cancer. DESIGN: This review was designed by searching words like cancer, oral cancer, cisplatin-resistance, tumor microenvironment, aberrant signalings, and extracellular vesicles, overcoming strategies for cisplatin resistance in databases like PubMed, Google Scholar, web science, and Scopus. Data available in this review is from 2017 to 2021. RESULTS: After searching too much data, we found these 98 data appropriate for our review. From these data, we found that tumor microenvironment, extracellular vesicles, and altered signaling pathways like PI3K/AKT, EGFR, NOTCH, Ras, PTEN, Nf-κß, and Wnt signaling have a crucial role in resistance development towards cisplatin in oral cancer. CONCLUSIONS: Lastly, this review explores the alternative strategies to overcome cisplatin resistance likely, the combination therapy and targeted therapy by combining more than one chemotherapeutic drug or inhibitors of signaling pathways and also by using nanoparticle loaded drugs that will reduce the drug efflux, which gives new treatment strategies.


Asunto(s)
Antineoplásicos , Neoplasias de la Boca , Humanos , Cisplatino/farmacología , Microambiente Tumoral , Fosfatidilinositol 3-Quinasas/metabolismo , Resistencia a Antineoplásicos , Vía de Señalización Wnt , Neoplasias de la Boca/tratamiento farmacológico , Antineoplásicos/farmacología , Línea Celular Tumoral
10.
Biochim Biophys Acta Rev Cancer ; 1875(2): 188517, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33524528

RESUMEN

Dysregulation of transcription factors is one of the common problems in the pathogenesis of human cancer. Among them, SOX9 is one of the critical transcription factors involved in various diseases, including cancer. The expression of SOX9 is regulated by microRNAs (miRNAs), methylation, phosphorylation, and acetylation. Interestingly, SOX9 acts as a proto-oncogene or tumor suppressor gene, relying upon kinds of cancer. Recent studies have reported the critical role of SOX9 in the regulation of the tumor microenvironment (TME). Additionally, activation of SOX9 signaling or SOX9 regulated signaling pathways play a crucial role in cancer development and progression. Accumulating evidence also suggests that SOX9 acquires stem cell features to induce epithelial-mesenchymal transition (EMT). Moreover, SOX9 has been broadly studied in the field of cancer stem cell (CSC) and EMT in the last decades. However, the link between SOX9 and cancer drug resistance has only recently been discovered. Furthermore, its differential expression could be a potential biomarker for tumor prognosis and progression. This review outlined the various biological implications of SOX9 in cancer progression and cancer drug resistance and elucidated its signaling network, which could be a potential target for designing novel anticancer drugs.


Asunto(s)
Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Factor de Transcripción SOX9/metabolismo , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , Humanos , Proto-Oncogenes Mas , Transducción de Señal
11.
Toxicol In Vitro ; 65: 104788, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32027944

RESUMEN

Prognosis of pancreatic cancer patients remains extremely poor thus, the need for the development of new therapeutic options is crucial. Plumbagin, a naphthoquinone derivative from Plumbago indica has been found to possess various pharmacological properties including anticancer activity. The present study was designed to investigate the inhibitory potential of plumbagin and associated mechanisms in pancreatic cancer cells. Fluorescence and flow cytometric analysis exhibited an increased percentage of apoptotic cells in both monolayer culture and 3D tumor spheroids. Upon plumbagin treatment, reactive oxygen species content of the cancer cells escalated and prompted alleviation of the mitochondrial membrane potential, which triggers caspase-dependent apoptosis. Interestingly, N-acetylcysteine inhibited the plumbagin induced apoptosis. We also found that the expression of Bcl-2 protein decreased and the expression of Bax protein increased. Moreover, plumbagin treatment led to upregulation of cleaved caspase-3 and caspase-9. These results support the views that plumbagin induced stress signals by damaging mitochondria and induce ROS mediated apoptosis via intrinsic apoptotic signaling in pancreatic cancer cells. To summarize, our study suggests that plumbagin may be utilized as a future anti-cancer therapy agent against pancreatic cancer, which is a major threat owing to its stubborn intransigence towards current treatment regimens.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Naftoquinonas/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Daño del ADN , Humanos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Mitocondrias/fisiología , Estrés Oxidativo/efectos de los fármacos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
12.
Food Chem Toxicol ; 125: 566-582, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30685472

RESUMEN

Plumbagin is a naphthoquinone derived yellow crystalline phytochemical. Plumbagin has a wide range of biological effects including cytotoxicity against cancer cells both in vitro and in vivo. Due to the pleiotropic nature of plumbagin, it shows the anticancer effect by targeting several molecular mechanisms including apoptosis and autophagic pathways, cell cycle arrest, anti-angiogenic pathways, anti-invasion and anti-metastasis pathways. Among many signaling pathways the key regulatory genes regulated by plumbagin are NF-kß, STAT3, and AKT, etc. Plumbagin is also a potent inducer of ROS, suppressor of cellular glutathione, and causes DNA strand break by oxidative DNA base damages. In vivo studies suggested that plumbagin significantly reduces the tumor weight and volume in dose-dependent manner without any side effects in tested model organisms. Another exciting aspect of plumbagin is the ability to re-sensitize the chemo and radioresistant cancer cells when used in combination or alone. Nano encapsulation of plumbagin overcomes the poor water solubility and bioavailability obstacles, enhancing the pharmaceutical relevance with better therapeutic efficacy. Moreover, plumbagin can be introduced as a future phytotherapeutic anticancer drug after fully satisfied preclinical and clinical trials.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Naftoquinonas/uso terapéutico , Neoplasias/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/toxicidad , Apoptosis/efectos de los fármacos , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Masculino , Naftoquinonas/farmacología , Naftoquinonas/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA