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










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(12)2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35743115

RESUMEN

In the fight against prostate cancer (PCa), TRPM8 is one of the most promising clinical targets. Indeed, several studies have highlighted that TRPM8 involvement is key in PCa progression because of its impact on cell proliferation, viability, and migration. However, data from the literature are somewhat contradictory regarding the precise role of TRPM8 in prostatic carcinogenesis and are mostly based on in vitro studies. The purpose of this study was to clarify the role played by TRPM8 in PCa progression. We used a prostate orthotopic xenograft mouse model to show that TRPM8 overexpression dramatically limited tumor growth and metastasis dissemination in vivo. Mechanistically, our in vitro data revealed that TRPM8 inhibited tumor growth by affecting the cell proliferation and clonogenic properties of PCa cells. Moreover, TRPM8 impacted metastatic dissemination mainly by impairing cytoskeleton dynamics and focal adhesion formation through the inhibition of the Cdc42, Rac1, ERK, and FAK pathways. Lastly, we proved the in vivo efficiency of a new tool based on lipid nanocapsules containing WS12 in limiting the TRPM8-positive cells' dissemination at metastatic sites. Our work strongly supports the protective role of TRPM8 on PCa progression, providing new insights into the potential application of TRPM8 as a therapeutic target in PCa treatment.


Asunto(s)
Neoplasias de la Próstata , Canales Catiónicos TRPM , Animales , Carcinogénesis/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Metástasis de la Neoplasia/patología , Próstata/patología , Neoplasias de la Próstata/metabolismo , Canales Catiónicos TRPM/genética , Canales Catiónicos TRPM/metabolismo
2.
Cell Death Dis ; 10(9): 652, 2019 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-31501416

RESUMEN

In prostate carcinogenesis, androgens are known to control the expression of the transient receptor potential melastatin 8 (TRPM8) protein via activation of androgen receptor (AR). Overexpression and/or activity of TRPM8 channel was shown to suppress prostate cancer (PCa) cell migration. Here we report that at certain concentrations androgens facilitate PCa cell migration. We show that underlying mechanism is inhibition of TRPM8 by activated AR which interacts with the channel within lipid rafts microdomains of the plasma membrane. Thus, our study has identified an additional nongenomic mechanism of the TRPM8 channel regulation by androgens that should be taken into account upon the development of novel therapeutic strategies.


Asunto(s)
Movimiento Celular/fisiología , Microdominios de Membrana/metabolismo , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/metabolismo , Canales Catiónicos TRPM/metabolismo , Biotinilación , Western Blotting , Movimiento Celular/genética , Silenciador del Gen/fisiología , Humanos , Inmunoprecipitación , Masculino , Células PC-3 , Neoplasias de la Próstata/genética , Receptores Androgénicos/genética , Imagen de Lapso de Tiempo , Análisis de Matrices Tisulares
3.
Methods Mol Biol ; 1557: 79-93, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28078584

RESUMEN

Intracellular calcium signals regulate cell function and cell survival by controlling many processes. CD95 engagement results in distinct intracellular calcium signals that control the cell fate, apoptosis, or survival, depending on the ligand (membrane or soluble). Intracellular calcium determination is an exquisite readout to explore the molecular mechanisms elicited by CD95 engagement. The most widely applied methods for studying calcium signaling pathways use fluorescent indicators and imaging methods with fluorescence microscopy. This technical approach, however, requires many precautions that we discuss in this chapter.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Receptor fas/metabolismo , Línea Celular , Proteína Ligando Fas/metabolismo , Humanos , Microscopía Confocal , Mitocondrias/metabolismo , Imagen Molecular/métodos , Unión Proteica
4.
Oncotarget ; 8(2): 3181-3196, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27911858

RESUMEN

Selective Serotonin Reuptake Inhibitor antidepressants, such as fluoxetine (Prozac), have been shown to induce cell death in cancer cells, paving the way for their potential use as cancer therapy. These compounds are able to increase cytosolic calcium concentration ([Ca2+]cyt), but the involved mechanisms and their physiological consequences are still not well understood. Here, we show that fluoxetine induces an increase in [Ca2+]cyt by emptying the endoplasmic reticulum (ER) through the translocon, an ER Ca2+ leakage structure. Our data also show that fluoxetine inhibits oxygen consumption and lowers mitochondrial ATP. This latter is essential for Ca2+ reuptake into the ER, and we postulated therefore that the fluoxetine-induced decrease in mitochondrial ATP production results in the emptying of the ER, leading to capacitative calcium entry. Furthermore, Ca2+ quickly accumulated in the mitochondria, leading to mitochondrial Ca2+ overload and cell death. We found that fluoxetine could induce an early necrosis in human peripheral blood lymphocytes and Jurkat cells, and could also induce late apoptosis, especially in the tumor cell line. These results shed light on fluoxetine-induced cell death and its potential use in cancer treatment.


Asunto(s)
Antidepresivos de Segunda Generación/farmacología , Calcio/metabolismo , Metabolismo Energético/efectos de los fármacos , Fluoxetina/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Necrosis/inducido químicamente , Necrosis/metabolismo , Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Inositol 1,4,5-Trifosfato/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Imagen Molecular , Consumo de Oxígeno , Fosfoinositido Fosfolipasa C/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Int J Cancer ; 137(6): 1306-17, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25704182

RESUMEN

Epigenomic changes are an important feature of malignant tumors. How tumor aggressiveness is affected by DNA methylation of specific loci is largely unexplored. In genome-wide DNA methylation analyses, we identified the KCa 3.1 channel gene (KCNN4) promoter to be hypomethylated in an aggressive non-small-cell lung carcinoma (NSCLC) cell line and in patient samples. Accordingly, KCa 3.1 expression was increased in more aggressive NSCLC cells. Both findings were strong predictors for poor prognosis in lung adenocarcinoma. Increased KCa 3.1 expression was associated with aggressive features of NSCLC cells. Proliferation and migration of pro-metastatic NSCLC cells depended on KCa 3.1 activity. Mechanistically, elevated KCa 3.1 expression hyperpolarized the membrane potential, thereby augmenting the driving force for Ca(2+) influx. KCa 3.1 blockade strongly reduced the growth of xenografted NSCLC cells in mice as measured by positron emission tomography-computed tomography. Thus, loss of DNA methylation of the KCNN4 promoter and increased KCa 3.1 channel expression and function are mechanistically linked to poor survival of NSCLC patients.


Asunto(s)
Epigénesis Genética/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Neoplasias Pulmonares/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenocarcinoma del Pulmón , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Metilación de ADN/genética , Epigenómica/métodos , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Pronóstico , Regiones Promotoras Genéticas/genética
6.
FASEB J ; 27(4): 1600-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23322163

RESUMEN

The endoplasmic reticulum (ER) is involved in many cellular functions, including protein folding and Ca(2+) homeostasis. The ability of cells to respond to the ER stress is critical for cell survival, and disruption in such regulation can lead to apoptosis. ER stress is accompanied by alterations in Ca(2+) homeostasis, and the ER Ca(2+) store depletion by itself can induce ER stress and apoptosis. Despite that, the ER Ca(2+) leak channels activated in response to the ER stress remain poorly characterized. Here we demonstrate that ER Ca(2+) depletion during the ER stress occurs via translocon, the ER protein complex involved in translation. Numerous ER stress inducers stimulate the ER Ca(2+) leak that can be prevented by translocon inhibitor, anisomycin. Expression of GRP78, an ER stress marker, increased following treatment with puromycin (a translocon opener) and was suppressed by anisomycin, confirming a primary role of translocon in ER stress induction. Inhibition of ER store depletion by anisomycin significantly reduces apoptosis stimulated by the ER stress inducers. We suggest that translocon opening is physiologically modulated by GRP78, particularly during the ER stress. The ability to modulate the ER Ca(2+) permeability and subsequent ER stress can lead to development of a novel therapeutic approach.


Asunto(s)
Apoptosis/fisiología , Calcio/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Retículo Endoplásmico/metabolismo , Proteínas de Choque Térmico/metabolismo , Respuesta de Proteína Desplegada , Anisomicina/farmacología , Canales de Calcio/efectos de los fármacos , Canales de Calcio/metabolismo , Células Cultivadas , Retículo Endoplásmico/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Homeostasis/fisiología , Humanos , Puromicina/farmacología , Respuesta de Proteína Desplegada/fisiología
7.
J Cell Physiol ; 227(12): 3837-46, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22495877

RESUMEN

Breast cancer (BC) has a poor prognosis due to its strong metastatic ability. Accumulating data present ether à go-go (hEag1) K(+) channels as relevant player in controlling cell cycle and proliferation of non-invasive BC cells. However, the role of hEag1 in invasive BC cells migration is still unknown. In this study, we studied both the functional expression and the involvement in cell migration of hEag1 in the highly metastatic MDA-MB-231 human BC cells. We showed that hEag1 mRNA and proteins were expressed in human invasive ductal carcinoma tissues and BC cell lines. Functional activity of hEag1 channels in MDA-MB-231 cells was confirmed using astemizole, a hEag1 blocker, or siRNA. Blocking or silencing hEag1 depolarized the membrane potential and reduced both Ca(2+) entry and MDA-MB-231 cell migration without affecting cell proliferation. Recent studies have reported that Ca(2+) entry through Orai1 channels is required for MDA-MB-231 cell migration. Down-regulation of hEag1 or Orai1 reduced Ca(2+) influx and cell migration with similar efficiency. Interestingly, no additive effects on Ca(2+) influx or cell migration were observed in cells co-transfected with sihEag1 and siOrai1. Finally, both Orai1 and hEag1 are expressed in invasive breast adenocarcinoma tissues and invaded metastatic lymph node samples (LNM(+)). In conclusion, this study is the first to demonstrate that hEag1 channels are involved in the serum-induced migration of BC cells by controlling the Ca(2+) entry through Orai1 channels. hEag1 may therefore represent a potential target for the suppression of BC cell migration, and thus prevention of metastasis development.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Movimiento Celular/fisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Canales de Calcio/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patología , Línea Celular Tumoral , Supervivencia Celular , Canales de Potasio Éter-A-Go-Go/genética , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , Inmunohistoquímica , Ganglios Linfáticos/patología , Manganeso , Invasividad Neoplásica , Proteína ORAI1 , Técnicas de Placa-Clamp , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...