Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO J ; 42(13): e112198, 2023 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-37278161

RESUMO

There is growing evidence that ion channels are critically involved in cancer cell invasiveness and metastasis. However, the molecular mechanisms of ion signaling promoting cancer behavior are poorly understood and the complexity of the underlying remodeling during metastasis remains to be explored. Here, using a variety of in vitro and in vivo techniques, we show that metastatic prostate cancer cells acquire a specific Na+ /Ca2+ signature required for persistent invasion. We identify the Na+ leak channel, NALCN, which is overexpressed in metastatic prostate cancer, as a major initiator and regulator of Ca2+ oscillations required for invadopodia formation. Indeed, NALCN-mediated Na+ influx into cancer cells maintains intracellular Ca2+ oscillations via a specific chain of ion transport proteins including plasmalemmal and mitochondrial Na+ /Ca2+ exchangers, SERCA and store-operated channels. This signaling cascade promotes activity of the NACLN-colocalized proto-oncogene Src kinase, actin remodeling and secretion of proteolytic enzymes, thus increasing cancer cell invasive potential and metastatic lesions in vivo. Overall, our findings provide new insights into an ion signaling pathway specific for metastatic cells where NALCN acts as persistent invasion controller.


Assuntos
Neoplasias da Próstata , Sódio , Masculino , Humanos , Sódio/metabolismo , Canais Iônicos/metabolismo , Transporte de Íons , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo
2.
Int J Mol Sci ; 20(24)2019 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-31888223

RESUMO

The characterization of calcium channel interactome in the last decades opened a new way of perceiving ion channel function and regulation. Partner proteins of ion channels can now be considered as major components of the calcium homeostatic mechanisms, while the reinforcement or disruption of their interaction with the channel units now represents an attractive target in research and therapeutics. In this review we will focus on the targeting of calcium channel partner proteins in order to act on the channel activity, and on its consequences for cell and organism physiology. Given the recent advances in the partner proteins' identification, characterization, as well as in the resolution of their interaction domain structures, we will develop the latest findings on the interacting proteins of the following channels: voltage-dependent calcium channels, transient receptor potential and ORAI channels, and inositol 1,4,5-trisphosphate receptor.


Assuntos
Bloqueadores dos Canais de Cálcio/uso terapêutico , Canais de Cálcio/metabolismo , Terapia de Alvo Molecular , Animais , Cálcio/metabolismo , Humanos , Modelos Biológicos , Ligação Proteica
3.
Mol Carcinog ; 56(8): 1851-1867, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28277613

RESUMO

Previous studies showed the effects of resveratrol (RES) on several cancer cells, including prostate cancer (PCa) cell apoptosis without taking into consideration the impact of the tumor microenvironment (TME). The TME is composed of cancer cells, endothelial cells, blood cells, and cancer-associated fibroblasts (CAF), the main source of growth factors. The latter cells might modify in the TME the impact of RES on tumor cells via secreted factors. Recent data clearly show the impact of CAF on cancer cells apoptosis resistance via secreted factors. However, the effects of RES on PCa CAF have not been studied so far. We have investigated here for the first time the effects of RES on the physiology of PCa CAF in the context of TME. Using a prostate cancer CAF cell line and primary cultures of CAF from prostate cancers, we show that RES activates the N-terminal mutated Transient Receptor Potential Ankyrin 1 (TRPA1) channel leading to an increase in intracellular calcium concentration and the expression and secretion of growth factors (HGF and VEGF) without inducing apoptosis in these cells. Interestingly, in the present work, we also show that when the prostate cancer cells were co-cultured with CAF, the RES-induced cancer cell apoptosis was reduced by 40%, an apoptosis reduction canceled in the presence of the TRPA1 channel inhibitors. The present work highlights CAF TRPA1 ion channels as a target for RES and the importance of the channel in the epithelial-stromal crosstalk in the TME leading to resistance to the RES-induced apoptosis.


Assuntos
Anticarcinógenos/farmacologia , Antioxidantes/farmacologia , Canais de Cálcio/metabolismo , Fibroblastos Associados a Câncer/efeitos dos fármacos , Proteínas do Tecido Nervoso/metabolismo , Próstata/efeitos dos fármacos , Neoplasias da Próstata/tratamento farmacológico , Estilbenos/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Canais de Cálcio/análise , Canais de Cálcio/genética , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Linhagem Celular Tumoral , Humanos , Masculino , Mutação , Proteínas do Tecido Nervoso/análise , Proteínas do Tecido Nervoso/genética , Próstata/metabolismo , Próstata/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Resveratrol , Canal de Cátion TRPA1 , Canais de Potencial de Receptor Transitório/análise , Canais de Potencial de Receptor Transitório/genética , Microambiente Tumoral/efeitos dos fármacos
4.
Eur Biophys J ; 45(7): 765-777, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27660079

RESUMO

Sodium (Na+) ions are known to regulate many signaling pathways involved in both physiological and pathological conditions. In particular, alterations in intracellular concentrations of Na+ and corresponding changes in membrane potential are known to be major actors of cancer progression to metastatic phenotype. Though the functionality of Na+ channels and the corresponding Na+ currents can be investigated using the patch-clamp technique, the latter is rather invasive and a technically difficult method to study intracellular Na+ transients compared to Na+ fluorescence imaging. Despite the fact that Na+ signaling is considered an important controller of cancer progression, only few data using Na+ imaging approaches are available so far, suggesting the persisting challenge within the scientific community. In this study, we describe in detail the approach for application of Na+ imaging technique to measure intracellular Na+ variations in human prostate cancer cells. Accordingly, we used three Na+-specific fluorescent dyes-Na+-binding benzofuran isophthalate (SBFI), CoroNa™ Green (Corona) and Asante NaTRIUM Green-2 (ANG-2). These dyes have been assessed for optimal loading conditions, dissociation constant and working range after different calibration methods, and intracellular Na+ sensitivity, in order to determine which probe can be considered as the most reliable to visualize Na+ fluctuations in vitro.


Assuntos
Corantes Fluorescentes/metabolismo , Espaço Intracelular/metabolismo , Imagem Molecular/métodos , Neoplasias da Próstata/patologia , Sódio/metabolismo , Calibragem , Linhagem Celular Tumoral , Citosol/efeitos dos fármacos , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Espaço Extracelular/efeitos dos fármacos , Espaço Extracelular/metabolismo , Humanos , Espaço Intracelular/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Masculino , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Canais de Sódio Disparados por Voltagem/metabolismo
5.
Proc Natl Acad Sci U S A ; 110(17): 6859-64, 2013 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-23569283

RESUMO

Defects in the human protein TMEM165 are known to cause a subtype of Congenital Disorders of Glycosylation. Transmembrane protein 165 (TMEM165) belongs to an uncharacterized family of membrane proteins called Uncharacterized Protein Family 0016, which are well conserved throughout evolution and share characteristics reminiscent of the cation/Ca(2+) exchanger superfamily. Gcr1 dependent translation factor 1 (Gdt1p), the budding yeast member of this family, contributes to Ca(2+) homeostasis via an uncharacterized Ca(2+) transport pathway localized in the Golgi apparatus. The gdt1Δ mutant was found to be sensitive to high concentrations of Ca(2+), and interestingly, this sensitivity was suppressed by expression of TMEM165, the human ortholog of Gdt1p, indicating conservation of function among the members of this family. Patch-clamp analyses on human cells indicated that TMEM165 expression is linked to Ca(2+) ion transport. Furthermore, defects in TMEM165 affected both Ca(2+) and pH homeostasis. Based on these results, we propose that Gdt1p and TMEM165 could be members of a unique family of Golgi-localized Ca(2+)/H(+) antiporters and that modification of the Golgi Ca(2+) and pH balance could explain the glycosylation defects observed in TMEM165-deficient patients.


Assuntos
Antiporters/metabolismo , Cálcio/metabolismo , Regulação da Expressão Gênica/fisiologia , Complexo de Golgi/metabolismo , Homeostase/fisiologia , Proteínas de Membrana/metabolismo , Saccharomycetales/metabolismo , Western Blotting , Proteínas de Transporte de Cátions , Fracionamento Celular , Citometria de Fluxo , Imunofluorescência , Regulação da Expressão Gênica/genética , Células HeLa , Humanos , Hidrogênio/metabolismo , Concentração de Íons de Hidrogênio , Técnicas de Patch-Clamp , RNA Interferente Pequeno/genética
6.
Exp Cell Res ; 315(2): 357-69, 2009 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19026635

RESUMO

Nucleolin is an ubiquitous nucleolar phosphoprotein involved in fundamental aspects of transcription regulation, cell proliferation and growth. It has also been described as a shuttling molecule between nucleus, cytosol and the cell surface. Several studies have demonstrated that surface nucleolin serves as a receptor for various extracellular ligands implicated in cell proliferation, differentiation, adhesion, mitogenesis and angiogenesis. Previously, we reported that nucleolin in the extranuclear cell compartment is a glycoprotein containing N- and O-glycans. In the present study, we show that glycosylation is an essential requirement for surface nucleolin expression, since it is prevented when cells are cultured in the presence of tunicamycin, an inhibitor of N-glycosylation. Accordingly, surface but not nuclear nucleolin is radioactively labeled upon metabolic labeling of cells with [(3)H]glucosamine. Besides its well-demonstrated role in the internalization of specific ligands, here we show that ligand binding to surface nucleolin could also induce Ca(2+) entry into cells. Indeed, by flow cytometry, microscopy and patch-clamp experiments, we show that the HB-19 pseudopeptide, which binds specifically surface nucleolin, triggers rapid and intense membrane Ca(2+) fluxes in various types of cells. The use of several drugs then indicated that Store-Operated Ca(2+) Entry (SOCE)-like channels are involved in the generation of these fluxes. Taken together, our findings suggest that binding of an extracellular ligand to surface nucleolin could be involved in the activation of signaling pathways by promoting Ca(2+) entry into cells.


Assuntos
Cálcio/metabolismo , Glicoproteínas/fisiologia , Fosfoproteínas/fisiologia , Proteínas de Ligação a RNA/fisiologia , Anticorpos/imunologia , Anticorpos/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Complexo CD3/imunologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio/fisiologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Proliferação de Células/efeitos dos fármacos , Citoplasma/metabolismo , Ácido Egtázico/farmacologia , Glucosamina/metabolismo , Glicoproteínas/antagonistas & inibidores , Glicoproteínas/biossíntese , Glicosilação/efeitos dos fármacos , Humanos , Células Jurkat , Técnicas de Patch-Clamp , Peptídeos/farmacologia , Fosfoproteínas/antagonistas & inibidores , Fosfoproteínas/biossíntese , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/biossíntese , Tunicamicina/farmacologia , Nucleolina
7.
Cancer Res ; 66(4): 2038-47, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16489003

RESUMO

One major clinical problem with prostate cancer is the cells' ability to survive and proliferate upon androgen withdrawal. Because Ca2+ is central to growth control, understanding the mechanisms of Ca2+ homeostasis involved in prostate cancer cell proliferation is imperative for new therapeutic strategies. Here, we show that agonist-mediated stimulation of alpha1-adrenergic receptors (alpha1-AR) promotes proliferation of the primary human prostate cancer epithelial (hPCE) cells by inducing store-independent Ca2+ entry and subsequent activation of nuclear factor of activated T cells (NFAT) transcription factor. Such an agonist-induced Ca2+ entry (ACE) relied mostly on transient receptor potential canonical 6 (TRPC6) channels, whose silencing by antisense hybrid depletion decreased both hPCE cell proliferation and ACE. In contrast, ACE and related growth arrest associated with purinergic receptors (P2Y-R) stimulation involved neither TRPC6 nor NFAT. Our findings show that alpha1-AR signaling requires the coupled activation of TRPC6 channels and NFAT to promote proliferation of hPCE cells and thereby suggest TRPC6 as a novel potential therapeutic target.


Assuntos
Cálcio/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Canais de Potencial de Receptor Transitório/metabolismo , Trifosfato de Adenosina/farmacologia , Agonistas de Receptores Adrenérgicos alfa 1 , Agonistas alfa-Adrenérgicos/farmacologia , Sinalização do Cálcio/fisiologia , Processos de Crescimento Celular/fisiologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Masculino , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fenilefrina/farmacologia , Receptores Purinérgicos/metabolismo , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6 , Regulação para Cima
8.
Cancer Prev Res (Phila) ; 10(3): 177-187, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28096238

RESUMO

Accruing evidence indicates that exposure to environmental compounds may adversely affect human health and promote carcinogenesis. Triclosan (TCS), an antimicrobial agent widely used as a preservative in personal care products, has been shown to act as an endocrine disruptor in hormone-dependent tissues. Here, we demonstrate a new molecular mechanism by which TCS stimulates the secretion by human prostate cancer stromal cells of vascular endothelial growth factor (VEGF), a factor known to promote tumor growth. This mechanism involves an increase in intracellular calcium levels due to the direct activation of a membrane ion channel. Using calcium imaging and electrophysiology techniques, we show for the first time that environmentally relevant concentrations of TCS activate a cation channel of the TRP family, TRPA1 (Transient Receptor Potential Ankirin 1), in primary cultured human prostate cancer stromal cells. The TCS-induced TRPA1 activation increased basal calcium in stromal cells and stimulated the secretion of VEGF and epithelial cells proliferation. Interestingly, immunofluorescence labeling performed on formalin-fixed paraffin-embedded prostate tissues showed an exclusive expression of the TRPA1 channel in prostate cancer stromal cells. Our data demonstrate an impact of the environmental factor TCS on the tumor microenvironment interactions, by activating a tumor stroma-specific TRPA1 ion channel. Cancer Prev Res; 10(3); 177-87. ©2017 AACR.


Assuntos
Anti-Infecciosos Locais/toxicidade , Canais de Cálcio/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Neoplasias da Próstata/metabolismo , Células Estromais/efeitos dos fármacos , Canais de Potencial de Receptor Transitório/metabolismo , Triclosan/toxicidade , Fator A de Crescimento do Endotélio Vascular/metabolismo , Carcinógenos Ambientais/toxicidade , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Células Estromais/metabolismo , Canal de Cátion TRPA1 , Microambiente Tumoral/efeitos dos fármacos
9.
Cell Calcium ; 39(4): 357-66, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16442617

RESUMO

Calcium entry through plasma membrane calcium channels is one of the most important cell signaling mechanism involved in such diverse functions as secretion, contraction and cell growth by regulating gene expression, proliferation and apoptosis. The identity of plasma membrane calcium channels, the main regulators of calcium entry, involved in cell proliferation has been thus extensively sought. Among these, a calcium entry pathway called capacitative calcium entry (CCE), activated by calcium store depletion, is particularly important in non-excitable cells. Though this capacitative calcium entry is generally supposed to occur through TRP channels there is some evidence that voltage-dependent T-type calcium channels may contribute to calcium entry after store depletion. Here we show that though mibefradil, a T-type calcium channel blocker, is able to reduce capacitative calcium entry induced by either thapsigargin or ATP, this was not mimicked by any other T-type calcium channel inhibitors even in cells overexpressing alpha(1H) T-type calcium channels, leading us to conclude that T-type calcium channels are not responsible for the capacitative calcium entry observed in different cancer cell lines. On the contrary, we show that the action of mibefradil on capacitative calcium entry is due to an action on store-operated calcium channels.


Assuntos
Canais de Cálcio Tipo T/fisiologia , Sinalização do Cálcio , Cálcio/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Ativação do Canal Iônico/fisiologia , Bloqueadores dos Canais de Cálcio/farmacologia , Canais de Cálcio , Canais de Cálcio Tipo T/efeitos dos fármacos , Canais de Cálcio Tipo T/metabolismo , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Transporte de Íons/fisiologia , Mibefradil/farmacologia , RNA Mensageiro/metabolismo , Transfecção
10.
Oncotarget ; 7(20): 29063-80, 2016 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-27074561

RESUMO

Since its cloning a decade ago, TRPM8 channel has emerged as a promising prognostic marker and a putative therapeutic target in prostate cancer (PCa). However, recent studies have brought to light the complexity of TRPM8 isoforms in PCa. Consequently, the respective role of each TRPM8 isoform needs to be deciphered prior to considering TRPM8 as an attractive therapeutic target. Full-length (6 transmembrane (TM)-domain) TRPM8 channel is overexpressed in early PCa and repressed in advanced prostate tumors whereas the localization of the truncated, 4TM-TRPM8 channel (4 transmembrane (TM)-domain), in the membranes of endoplasmic reticulum (ER) is independent of the pathogenic status of epithelial cells. In the same line, expression of non-channel cytoplasmic small TRPM8 isoforms (namely sM8) is conserved in cancer cells. In this study, we identify sM8s as putative regulator of PCa cell death. Indeed, suppression of sM8 isoforms was found to induce concomitantly ER stress, oxidative stress, p21 expression and apoptosis in human epithelial prostate cancer cells. We furthermore demonstrate that induction of such mechanisms required the activity of 4TM-TRPM8 channels at the ER-mitochondria junction. Our study thus suggests that targeting sM8 could be an appropriate strategy to fight prostate cancer.


Assuntos
Neoplasias da Próstata/patologia , Canais de Cátion TRPM/metabolismo , Animais , Apoptose/fisiologia , Linhagem Celular Tumoral , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias da Próstata/metabolismo , Isoformas de Proteínas/metabolismo
11.
Springerplus ; 2(1): 54, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23450760

RESUMO

Bisphenol A (BPA), the principal constituent of reusable water bottles, metal cans, and plastic food containers, has been shown to be involved in human prostate cancer (PCa) cell proliferation. The aim of the present study was to explore the effect of BPA on PCa cell migration and the pathways involved in these processes. Using the transwell technique, we clearly show for the first time that the pre-treatment of the cells with BPA (1-10 nM) induces human PCa cell migration. Using a calcium imaging technique, we show that BPA pre-treatment induces an amplification of Store-Operated Calcium Entry (SOCE) in LNCaP cells. RT-PCR and Western blot experiments allowed the identification of the ion channel proteins which are up-regulated by BPA pre-treatments. These include the Orai1 protein, which is known as an important SOCE actor in various cell systems, including human PCa cells. Using a siRNA strategy, we observed that BPA-induced amplification of SOCE was Orai1-dependent. Interestingly, the BPA-induced PCa cell migration was suppressed when the calcium entry was impaired by the use of SOCE inhibitors (SKF96365, BTP2), or when the extracellular calcium was chelated. Taken together, the results presented here show that BPA induces PCa cells migration via a modulation of the ion channel protein expression involved in calcium entry and in cancer cell migration. The present data provide novel insights into the molecular mechanisms involved in the effects of an environmental factor on cancer cells and suggest both the necessity of preventive measures and the possibility of targeting ion channels in the treatment of PCa cell metastasis.

12.
Biol Open ; 2(9): 941-51, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24143281

RESUMO

It is strongly suspected that potassium (K(+)) channels are involved in various aspects of prostate cancer development, such as cell growth. However, the molecular nature of those K(+) channels implicated in prostate cancer cell proliferation and the mechanisms through which they control proliferation are still unknown. This study uses pharmacological, biophysical and molecular approaches to show that the main voltage-dependent K(+) current in prostate cancer LNCaP cells is carried by large-conductance BK channels. Indeed, most of the voltage-dependent current was inhibited by inhibitors of BK channels (paxillin and iberiotoxin) and by siRNA targeting BK channels. In addition, we reveal that BK channels constitute the main K(+) channel family involved in setting the resting membrane potential in LNCaP cells at around -40 mV. This consequently promotes a constitutive calcium entry through T-type Cav3.2 calcium channels. We demonstrate, using single-channel recording, confocal imaging and co-immunoprecipitation approaches, that both channels form macromolecular complexes. Finally, using flow cytometry cell cycle measurements, cell survival assays and Ki67 immunofluorescent staining, we show that both BK and Cav3.2 channels participate in the proliferation of prostate cancer cells.

13.
PLoS One ; 7(9): e45615, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23049826

RESUMO

Neuroendocrine differentiation (NED) is a hallmark of advanced androgen-independent prostate cancer, for which no successful therapy exists. NED tumour cells escape apoptotic cell death by alterations of Ca(2+) homeostasis where the store-operated Ca(2+) entry (SOCE) is known to be a key event. We have previously shown that the downregulation of Orai1 protein representing the major molecular component of endogenous SOCE in human prostate cancer cells, and constituting the principal source of Ca(2+) influx used by the cell to trigger apoptosis, contributes to the establishment of an apoptosis-resistant phenotype (Cell Death Dis. 2010 Sep 16;1:e75.). Here, we report for the first time that the decrease of SOCE during NED may be caused by alternative NED-induced mechanism involving cytoskeleton reorganisation. NED induced by androgen deprivation resulted in a decrease of SOCE due to cortical F-actin over-polymerization which inhibits thapsigargin-induced SOCE. The disruption of F-actin polymerization by Cytochalasin D in NED cells restored SOCE, while the induction of F-actin polymerization by jasplakinolide or calyculin A diminished SOCE without changing the expression of key SOCE players: Orai1, STIM1, and TRPC1. Our data suggest that targeting cytoskeleton-induced pathways of malignant cells together with SOCE-involved channels may prove a useful strategy in the treatment of advanced prostate cancer.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Citoesqueleto/metabolismo , Células Neuroendócrinas/citologia , Actinas/metabolismo , Androgênios/metabolismo , Apoptose , Diferenciação Celular , Linhagem Celular Tumoral , Citocalasina D/farmacologia , Primers do DNA/genética , Eletrofisiologia/métodos , Humanos , Masculino , Toxinas Marinhas , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína ORAI1 , Oxazóis/farmacologia , Fenótipo , Neoplasias da Próstata/terapia , Molécula 1 de Interação Estromal , Canais de Cátion TRPC/metabolismo
14.
PLoS One ; 4(9): e7068, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19763272

RESUMO

BACKGROUND: During androgen ablation prostate cancer cells' growth and survival become independent of normal regulatory mechanisms. These androgen-independent cells acquire the remarkable ability to adapt to the surrounding microenvironment whose factors, such as neurotransmitters, influence their survival. Although findings are becoming evident about the expression of alpha(1A)-adrenoceptors in prostate cancer epithelial cells, their exact functional role in androgen-independent cells has yet to be established. Previous work has demonstrated that membrane lipid rafts associated with key signalling proteins mediate growth and survival signalling pathways in prostate cancer cells. METHODOLOGY/PRINCIPAL FINDINGS: In order to analyze the membrane topology of the alpha(1A)-adrenoceptor we explored its presence by a biochemical approach in purified detergent resistant membrane fractions of the androgen-independent prostate cancer cell line DU145. Electron microscopy observations demonstrated the colocalization of the alpha(1A)-adrenoceptor with caveolin-1, the major protein component of caveolae. In addition, we showed that agonist stimulation of the alpha(1A)-adrenoceptor induced resistance to thapsigargin-induced apoptosis and that caveolin-1 was necessary for this process. Further, immunohistofluorescence revealed the relation between high levels of alpha(1A)-adrenoceptor and caveolin-1 expression with advanced stage prostate cancer. We also show by immunoblotting that the TG-induced apoptosis resistance described in DU145 cells is mediated by extracellular signal-regulated kinases (ERK). CONCLUSIONS/SIGNIFICANCE: In conclusion, we propose that alpha(1A)-adrenoceptor stimulation in androgen-independent prostate cancer cells via caveolae constitutes one of the mechanisms contributing to their protection from TG-induced apoptosis.


Assuntos
Apoptose , Cavéolas/metabolismo , Resistencia a Medicamentos Antineoplásicos , Neoplasias da Próstata/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Colesterol/metabolismo , Humanos , Masculino , Microdomínios da Membrana , Modelos Biológicos , Neurotransmissores , Neoplasias da Próstata/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Transdução de Sinais , Esfingomielinas/metabolismo , Tapsigargina/farmacologia
15.
J Biol Chem ; 283(15): 10162-73, 2008 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-18230611

RESUMO

Because prostate cancer is, in its early stages, an androgen-dependent pathology, treatments aiming at decreasing testosterone plasma concentration have been developed for many years now. However, a significant proportion of patients suffer a relapse after a few years of hormone therapy. The androgen-independent stage of prostate cancer has been shown to be associated with the development of neuroendocrine differentiation. We previously demonstrated that neuroendocrine prostate cancer cells derived from LNCaP cells overexpress CaV3.2 T-type voltage-dependent calcium channels. We demonstrate here using prostatic acid phosphatase as a marker of prostate secretion and FM1-43 fluorescence imaging of membrane trafficking that neuroendocrine differentiation is associated with an increase in calcium-dependent secretion which critically relies on CaV3.2 T-type calcium channel activity. In addition, we show that these channels are expressed by neuroendocrine cells in prostate cancer tissues obtained from patients after surgery. We propose that CaV3.2 T-type calcium channel up-regulation may account for the alteration of secretion during prostate cancer development and that these channels, by promoting the secretion of potential mitogenic factors, could participate in the progression of the disease toward an androgen-independent stage.


Assuntos
Biomarcadores Tumorais/metabolismo , Canais de Cálcio Tipo T/metabolismo , Cálcio/metabolismo , Carcinoma Neuroendócrino/metabolismo , Substâncias de Crescimento/metabolismo , Neoplasias da Próstata/metabolismo , Fosfatase Ácida , Androgênios/sangue , Carcinoma Neuroendócrino/patologia , Carcinoma Neuroendócrino/terapia , Diferenciação Celular , Linhagem Celular Tumoral , Terapia de Reposição Hormonal , Humanos , Masculino , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Proteínas Tirosina Fosfatases/metabolismo , Testosterona/sangue , Regulação para Cima
16.
Mol Pharmacol ; 71(3): 843-51, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17164406

RESUMO

In the present study, we investigated the effect of the antiestrogen compound tamoxifen on BK channels by the use of the patch-clamp technique. The perfusion of 10 nM tamoxifen significantly increased the magnitude of a voltage-dependent K+ current by 22.6 +/- 10.6% (n = 23). The effect of tamoxifen was always obtained in the first minute, peaked at 5.9 +/- 2.2 min (n = 23), and was abolished by the perfusion of tetraethylammonium (0.5 mM), charybdotoxin (50 nM), or iberiotoxin (100 nM). The stimulatory effect of 10 nM tamoxifen was the same at low (50 nM) and high (700 nM) internal calcium concentration and was not additive to that of 17-beta-estradiol (E2) or its membrane-impermeant form, beta-estradiol 6-(O-carboxymethyl)oxime:bovine serum albumin. Furthermore, the effect of tamoxifen was still recorded in the presence of the selective estrogen receptor antagonist faslodex (ICI-182,780; 1 microM). At the single-channel level, tamoxifen significantly increased the open probability of the BK channel by 46.2 +/- 10.1% (n = 4) without changing its unitary conductance. Moreover, we show here that the stimulation of BK channel activity by tamoxifen is involved in MCF-7 cell proliferation. Taken together, these results permitted us to identify the BK channel as the molecular target of tamoxifen that probably acts at the same extracellular molecular level as E2. The site of action of tamoxifen is probably the channel itself or the auxiliary beta subunits.


Assuntos
Neoplasias da Mama/patologia , Antagonistas de Estrogênios/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Tamoxifeno/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Estradiol/análogos & derivados , Estradiol/farmacologia , Feminino , Fulvestranto , Humanos , Compostos de Tetraetilamônio/farmacologia
17.
J Biol Chem ; 277(13): 10824-33, 2002 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-11799114

RESUMO

Neuroendocrine differentiation of prostate epithelial cells is usually associated with an increased aggressivity and invasiveness of prostate tumors and a poor prognosis. However, the molecular mechanisms involved in this process remain poorly understood. We have investigated the possible expression of voltage-gated calcium channels in human prostate cancer epithelial LNCaP cells and their modulation during neuroendocrine differentiation. A small proportion of undifferentiated LNCaP cells displayed a voltage-dependent calcium current. This proportion and the calcium current density were significantly increased during neuroendocrine differentiation induced by long-term treatments with cyclic AMP permeant analogs or with a steroid-reduced culture medium. Biophysical and pharmacological properties of this calcium current suggest that it is carried by low-voltage activated T-type calcium channels. Reverse transcriptase-PCR experiments demonstrated that only a single type of LVA calcium channel mRNA, an alpha(1H) calcium channel mRNA, is expressed in LNCaP cells. Quantitative real-time reverse transcriptase-PCR revealed that alpha(1H) mRNA was overexpressed during neuroendocrine differentiation. Finally, we show that this calcium channel promotes basal calcium entry at resting membrane potential and may facilitate neurite lengthening. This voltage-dependent calcium channel could be involved in the stimulation of mitogenic factor secretion and could therefore be a target for future therapeutic strategies.


Assuntos
Canais de Cálcio Tipo T/genética , Carcinoma Neuroendócrino/metabolismo , Diferenciação Celular , Neoplasias da Próstata/metabolismo , Sequência de Bases , Carcinoma Neuroendócrino/fisiopatologia , Carcinoma Neuroendócrino/prevenção & controle , AMP Cíclico/metabolismo , Primers do DNA , Humanos , Masculino , Potenciais da Membrana , Neoplasias da Próstata/patologia , Neoplasias da Próstata/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA