Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-32761457

RESUMEN

In recent years, it has been shown that breast cancer consists not only of neoplastic cells, but also of significant alterations in the surrounding stroma or tumor microenvironment. These alterations are now recognized as a critical element for breast cancer development and progression, as well as potential therapeutic targets. Furthermore, there is no doubt that ion channels are deregulated in breast cancer and some of which are prognostic markers of clinical outcome. Their dysregulation is also associated with aberrant signaling pathways. The number of published data on ion channels modifications by the microenvironment has significantly increased last years. Here, we summarize the state of the art on the cross talk between the tumor microenvironment and ion channels, in particular collagen 1, EGF, TGF-ß, ATP, hypoxia, and pH, on the development and progression of breast cancer.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Canales Iónicos , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
2.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-36142596

RESUMEN

In the fight against cancer, therapeutic strategies using cisplatin are severely limited by the appearance of a resistant phenotype. While cisplatin is usually efficient at the beginning of the treatment, several patients endure resistance to this agent and face relapse. One of the reasons for this resistant phenotype is the emergence of a cell subpopulation known as cancer stem cells (CSCs). Due to their quiescent phenotype and self-renewal abilities, these cells have recently been recognized as a crucial field of investigation in cancer and treatment resistance. Changes in intracellular calcium (Ca2+) through Ca2+ channel activity are essential for many cellular processes such as proliferation, migration, differentiation, and survival in various cell types. It is now proved that altered Ca2+ signaling is a hallmark of cancer, and several Ca2+ channels have been linked to CSC functions and therapy resistance. Moreover, cisplatin was shown to interfere with Ca2+ homeostasis; thus, it is considered likely that cisplatin-induced aberrant Ca2+ signaling is linked to CSCs biology and, therefore, therapy failure. The molecular signature defining the resistant phenotype varies between tumors, and the number of resistance mechanisms activated in response to a range of pressures dictates the global degree of cisplatin resistance. However, if we can understand the molecular mechanisms linking Ca2+ to cisplatin-induced resistance and CSC behaviors, alternative and novel therapeutic strategies could be considered. In this review, we examine how cisplatin interferes with Ca2+ homeostasis in tumor cells. We also summarize how cisplatin induces CSC markers in cancer. Finally, we highlight the role of Ca2+ in cancer stemness and focus on how they are involved in cisplatin-induced resistance through the increase of cancer stem cell populations and via specific pathways.


Asunto(s)
Cisplatino , Neoplasias , Calcio/metabolismo , Línea Celular Tumoral , Cisplatino/metabolismo , Cisplatino/farmacología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Humanos , Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo
3.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-35887266

RESUMEN

Dysregulation of the transient receptor canonical ion channel (TRPC1) has been found in several cancer types, yet the underlying molecular mechanisms through which TRPC1 impacts pancreatic ductal adenocarcinoma (PDAC) cell proliferation are incompletely understood. Here, we found that TRPC1 is upregulated in human PDAC tissue compared to adjacent pancreatic tissue and this higher expression correlates with low overall survival. TRPC1 is, as well, upregulated in the aggressive PDAC cell line PANC-1, compared to a duct-like cell line, and its knockdown (KD) reduced cell proliferation along with PANC-1 3D spheroid growth by arresting cells in the G1/S phase whilst decreasing cyclin A, CDK2, CDK6, and increasing p21CIP1 expression. In addition, the KD of TRPC1 neither affected Ca2+ influx nor store-operated Ca2+ entry (SOCE) and reduced cell proliferation independently of extracellular calcium. Interestingly, TRPC1 interacted with the PI3K-p85α subunit and calmodulin (CaM); both the CaM protein level and AKT phosphorylation were reduced upon TRPC1 KD. In conclusion, our results show that TRPC1 regulates PDAC cell proliferation and cell cycle progression by interacting with PI3K-p85α and CaM through a Ca2+-independent pathway.


Asunto(s)
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Calcio/metabolismo , Calmodulina/metabolismo , Carcinoma Ductal Pancreático/genética , Proliferación Celular , Humanos , Neoplasias Pancreáticas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Canales Catiónicos TRPC/genética , Canales Catiónicos TRPC/metabolismo , Neoplasias Pancreáticas
4.
J Cell Physiol ; 232(12): 3631-3640, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28145580

RESUMEN

Aberrant glycosylation changes on many glycoproteins are often related to cancer progression and metastasis. sp2 -Iminosugar-type castanospermine analogues, inhibitors of α-glucosidases, have been reported to exhibit antitumor activity. However, their effects on cell migration and the underlying molecular mechanism are not fully understood. Here, we investigated the effect of the pseudo-C-octyl glycoside 2-oxa-3-oxocastanospermine derivatives (CO-OCS) on breast cancer cells (MCF-7 and MDA-MB-231 cells), and MCF-10A mammary normal cell lines. We showed that CO-OCS treatment results in the drastic decrease of breast cancer cell migration without affecting cell proliferation. Furthermore, CO-OCS significantly reduced both the expression of ß1-integrin, which is a crucial interacting partner of Focal Adhesion Kinase (FAK), and the phosphorylation rates of FAK and ERK1/2. CO-OCS also drastically reduced Ca2+ entry through Store Operated Channels (SOC). Orai1 and Stim1, two N-glycosylated proteins, are involved in Store-Operated Calcium Entry (SOCE), and are essential for breast tumor cell migration. Our results showed that CO-OCS decreased the expression, at the protein level, of Stim1 without affecting that of Orai1. Moreover, cell migration and SOCE were attenuated by CO-OCS as well as when Stim1 was silenced. In contrast, in MCF-10A cells, CO-OCS slightly reduced cell migration, but was without effect on gene expression of Stim1, Orai1, ß1-integrin, or FAK and ERK1/2 activation. Our results provide strong evidence for a significant effect of CO-OCS on breast cancer cell migration and support that this effect was associated with ß1-integrin, Stim1, and FAK signaling pathways.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Movimiento Celular/efectos de los fármacos , Quinasa 1 de Adhesión Focal/metabolismo , Inhibidores de Glicósido Hidrolasas/farmacología , Indolizinas/farmacocinética , Integrina beta1/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula de Interacción Estromal 1/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Glicosilación , Humanos , Células MCF-7 , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Molécula de Interacción Estromal 1/genética , Factores de Tiempo , Transfección
5.
Biochim Biophys Acta ; 1848(10 Pt B): 2621-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25703813

RESUMEN

DNA methylation at CpG sites is an epigenetic mechanism that regulates cellular gene expression. In cancer cells, aberrant methylation is correlated with the abnormalities in expression of genes that are known to be involved in the particular characteristics of cancer cells such as proliferation, apoptosis, migration or invasion. During the past 30 years, accumulating data have definitely convinced the scientific community that ion channels are involved in cancerogenesis and cancer properties. As they are situated at the cell surface, they might be prime targets in the development of new therapeutic strategies besides their potential use as prognostic factors. Despite the progress in our understanding of the remodeling of ion channels in cancer cells, the molecular mechanisms underlying their over- or down-expression remained enigmatic. In this review, we aimed to summarize the available data on gene promoter methylation of ion channels and to investigate their clinical significance as novel biomarkers in cancer. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Asunto(s)
Biomarcadores de Tumor/genética , ADN de Neoplasias/metabolismo , Epigénesis Genética , Neoplasias/diagnóstico , Neoplasias/genética , Biomarcadores de Tumor/metabolismo , Canales de Calcio/genética , Canales de Calcio/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Metilación de ADN , ADN de Neoplasias/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Canales de Potasio/genética , Canales de Potasio/metabolismo , Regiones Promotoras Genéticas , Receptores Ionotrópicos de Glutamato/genética , Receptores Ionotrópicos de Glutamato/metabolismo , Transducción de Señal , Células Tumorales Cultivadas
6.
Pflugers Arch ; 468(5): 751-62, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26743871

RESUMEN

K(+) ions play a major role in many cellular processes. The deregulation of K(+) signaling is associated with a variety of diseases including cancer. Ether-à-go-go-1 (Eag1, Kv10.1, KCNH1) is a member of the voltage-activated potassium channel family and was the first K(+) channel to be associated with oncogenesis and tumor development. Interestingly, in healthy tissue, Kv10.1 is only detected in the central nervous system, where it is involved in the regulation of excitability under repeated stimulation. Kv10.1 is in contrast robustly expressed in over 70 % human tumors, where its expression seems to be controlled by key regulators of proliferation and survival such as p53 and E2F1, often altered in cancer. Otherwise, Kv10.1 is involved in cell proliferation, survival, angiogenesis, migration, and invasion. This review aims to provide a comprehensive overview of the current status of research on the role of Kv10.1 channel in physiopathology. Focus is placed on biophysical and pharmacological properties of Kv10.1 channel, as well as its cycling, trafficking, and its role in the neuron and cancer. The possible mechanisms by which Kv10.1 channel affects tumor cell migration and survival in breast cancer and its regulation by extracellular proteins are discussed.


Asunto(s)
Carcinogénesis/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Microambiente Tumoral , Animales , Movimiento Celular , Proliferación Celular , Canales de Potasio Éter-A-Go-Go/genética , Humanos , Neuronas/metabolismo , Neuronas/fisiología
7.
Biochim Biophys Acta ; 1833(3): 752-60, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23266555

RESUMEN

Members of the Orai family are highly selective calcium ion channels that play an important role in store-operated calcium entry. Among the three known Orai isoforms, Orai3 has gained increased attention, notably for its emerging role in cancer. We recently demonstrated that Orai3 channels are over-expressed in breast cancer (BC) biopsies, and involved specifically in proliferation, cell cycle progression and survival of MCF-7 BC cells. Here, we investigate the downstream signaling mechanisms affected by Orai3 silencing, leading to the subsequent functional impact specifically seen in MCF-7 cancer cells. We report a correlation between Orai3 and c-myc expression in tumor tissues and in the MCF-7 cancer cell line by demonstrating that Orai3 down-regulation reduces both expression and activity of the proto-oncogene c-myc. This is likely mediated through the MAP Kinase pathway, as we observed decreased pERK1/2 levels and cell-cycle arrest in G1 phase after Orai3 silencing. Our results provide strong evidence that the c-myc proto-oncogene is influenced by the store-operated calcium entry channel Orai3 through the MAP kinase pathway. This connection provides new clues in the downstream mechanism linking Orai3 channels and proliferation, cell cycle progression and survival of MCF-7 BC cells.


Asunto(s)
Neoplasias de la Mama/patología , Canales de Calcio/metabolismo , Proliferación Celular , Fase G1/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Adenocarcinoma , Apoptosis , Western Blotting , Mama/citología , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Calcio/metabolismo , Canales de Calcio/química , Canales de Calcio/genética , Células Cultivadas , Femenino , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Técnicas para Inmunoenzimas , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-myc/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Análisis de Matrices Tisulares
8.
Cell Calcium ; 113: 102760, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37247443

RESUMEN

Breast cancer (BC) is the second most common cancer and cause of death in women. The aggressive subtypes including triple negative types (TNBCs) show a resistance to chemotherapy, impaired immune system, and a worse prognosis. From a histological point of view, TNBCs are deficient in oestrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (HER2+) expression. Many studies reported an alteration in the expression of calcium channels, calcium binding proteins and pumps in BC that promote proliferation, survival, resistance to chemotherapy, and metastasis. Moreover, Ca2+ signal remodeling and calcium transporters expression have been associated to TNBCs and HER2+ BC subtypes. This review provides insight into the underlying alteration of the expression of calcium-permeable channels, pumps, and calcium dependent proteins and how this alteration plays an important role in promoting metastasis, metabolic switching, inflammation, and escape to chemotherapy treatment and immune surveillance in aggressive BC including TNBCs models and highly metastatic BC tumors.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Neoplasias de la Mama/metabolismo , Calcio
9.
Cancers (Basel) ; 14(19)2022 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-36230869

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a low overall survival rate of less than 10% and limited therapeutic options. Fluctuations in tumor microenvironment pH are a hallmark of PDAC development and progression. Many ion channels are bona fide cellular sensors of changes in pH. Yet, the interplay between the acidic tumor microenvironment and ion channel regulation in PDAC is poorly understood. In this study, we show that acid adaption increases PANC-1 cell migration but attenuates proliferation and spheroid growth, which are restored upon recovery. Moreover, acid adaptation and recovery conditions favor the plasma membrane localization of the pH-sensitive calcium (Ca2+) channel transient receptor potential C1 (TRPC1), TRPC1-mediated Ca2+ influx, channel interaction with the PI3K p85α subunit and calmodulin (CaM), and AKT and ERK1/2 activation. Knockdown (KD) of TRPC1 suppresses cell migration, proliferation, and spheroid growth, notably in acid-recovered cells. KD of TRPC1 causes the accumulation of cells in G0/G1 and G2/M phases, along with reduced expression of CDK6, -2, and -1, and cyclin A, and increased expression of p21CIP1. TRPC1 silencing decreases the basal Ca2+ influx in acid-adapted and -recovered cells, but not in normal pH conditions, and Ca2+ chelation reduces cell migration and proliferation solely in acid adaptation and recovery conditions. In conclusion, acid adaptation and recovery reinforce the involvement of TRPC1 in migration, proliferation, and cell cycle progression by permitting Ca2+ entry and forming a complex with the PI3K p85α subunit and CaM.

10.
J Cell Physiol ; 226(2): 542-51, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20683915

RESUMEN

Breast cancer (BC) is the leading cancer in the world in terms of incidence and mortality in women. However, the mechanism by which BC develops remains largely unknown. The increase in cytosolic free Ca(2+) can result in different physiological changes including cell growth and death. Orai isoforms are highly Ca(2+) selective channels. In the present study, we analyzed Orai3 expression in normal and cancerous breast tissue samples, and its role in MCF-7 BC and normal MCF-10A mammary epithelial cell lines. We found that the expression of Orai3 mRNAs was higher in BC tissues and MCF-7 cells than in normal tissues and MCF-10A cells. Down-regulation of Orai3 by siRNA inhibited MCF-7 cell proliferation and arrested cell cycle at G1 phase. This phenomenon is associated with a reduction in CDKs 4/2 (cyclin-dependent kinases) and cyclins E and D1 expression and an accumulation of p21(Waf1/Cip1) (a cyclin-dependent kinase inhibitor) and p53 (a tumor-suppressing protein). Orai3 was also involved in MCF-7 cell survival. Furthermore, Orai3 mediated Ca(2+) entry and contributed to intracellular calcium concentration ([Ca(2+)](i)). In MCF-10A cells, silencing Orai3 failed to modify [Ca(2+)](i), cell proliferation, cell-cycle progression, cyclins (D1, E), CDKs (4, 2), and p21(Waf1/Cip1) expression. Our results provide strong evidence for a significant effect of Orai3 on BC cell growth in vitro and show that this effect is associated with the induction of cell cycle and apoptosis resistance. Our study highlights a possible role of Orai3 as therapeutic target in BC therapy.


Asunto(s)
Apoptosis/fisiología , Neoplasias de la Mama , Mama/citología , Canales de Calcio/metabolismo , Ciclo Celular/fisiología , Células Epiteliales/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Canales de Calcio/genética , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Regulación hacia Abajo , Células Epiteliales/citología , Femenino , Humanos , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
11.
Cell Physiol Biochem ; 28(5): 813-22, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22178934

RESUMEN

BACKGROUND: Transient Receptor Potential (TRP) channels are expressed in many solid tumors. However, their expression in breast cancer remains largely unknown. Here, we investigated the profile expression of 13 TRP channels in human breast ductal adenocarcinoma (hBDA) and performed a correlation between their overexpression and pathological parameters. METHODS: The TRP channels expression was determined by RT-PCR in hBDA tissue, in human breast cancer epithelial (hBCE) primary culture and in MCF-7 cell line. The TRP protein level was evaluated by immunohistochemistry in hBDA tissue samples of 59 patients. RESULTS: TRPC1, TRPC6, TRPM7, TRPM8, and TRPV6 channels were overexpressed in hBDA compared to the adjacent non-tumoral tissue. Most interestingly, TRPC1, TRPM7 and TRPM8 expression strongly correlated with proliferative parameters (SBR grade, Ki67 proliferation index, and tumor size), and TRPV6 was mainly overexpressed in the invasive breast cancer cells. Using laser capture microdissection, we found that TRPV6 expression was higher in invasive areas, compared to the corresponding non-invasive ones. Moreover, TRPV6 silencing inhibited MDA-MB-231 migration and invasion, and MCF-7 migration. CONCLUSION: TRP channels are aberrantly expressed in hBDA, hBCE primary cultures, and cell lines, and associated with pathological parameters. The high expression of TRP channels in tumors suggests the potential of these channels for diagnostic, prognosis and/or therapeutic approaches in human breast ductal adenocarcinoma.


Asunto(s)
Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Canales de Potencial de Receptor Transitorio/metabolismo , Neoplasias de la Mama/metabolismo , Células Cultivadas , Femenino , Humanos , Inmunohistoquímica , Estadificación de Neoplasias , Proteínas Serina-Treonina Quinasas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6 , Canales Catiónicos TRPM/metabolismo , Canales Catiónicos TRPV/antagonistas & inhibidores , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/genética
12.
J Membr Biol ; 234(1): 47-56, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20177667

RESUMEN

Prolactin (PRL) is a polypeptidic hormone which acts both systemically and locally to cause lactation by interacting with the PRL receptor, a Janus kinase (JAK2)-coupled cytokine receptor family member. Several studies have reported that serum PRL level elevation is associated with an increased risk for breast cancer, and evidence has suggested that PRL is one actor in the pathogenesis and progression of this cancer. We previously reported the involvement of hIKCa1 in breast cell cycle progression and cell proliferation. However, mechanisms by which PRL cooperates with these channels to modulate breast epithelial cell proliferation remain unknown. Our results showed that, in the MCF-7 breast cancer cell line, PRL increased hIKCa1 current density. These channels were functional and regulated the resting membrane potential. The PRL effects were inhibited by TRAM-34 and clotrimazole, the most used hIKCa1 blockers. Moreover, PRL increased proliferation in a dose-dependent manner without overexpressing hIKCa1. To determine whether PRL-induced proliferation and hIKCa1 activity involved the JAK2 pathway, we used pharmacological JAK2 inhibitors (AG490 and JAK inhibitor I). Indeed, PRL-induced JAK2 phosphorylation was required for both cell proliferation and hIKCa1 activity. In the presence of either hIKCa1 blockers or siRNA-hIKCa1, PRL failed to increase cell proliferation and hIKCa1 activity. Taken together, our results demonstrate that PRL plays a role in breast cancer cell proliferation by increasing hIKCa1 activity through the JAK2 signaling pathway.


Asunto(s)
Canales de Potasio de Conductancia Intermedia Activados por el Calcio/fisiología , Janus Quinasa 2/fisiología , Receptores de Prolactina/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Calcio/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Clotrimazol/farmacología , Femenino , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/efectos de los fármacos , Prolactina/farmacología , Pirazoles , Tirfostinos/farmacología
13.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118644, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31931022

RESUMEN

For twenty years, ion channels have been studied in cancer progression. Several information have been collected about their involvement in cancer cellular processes like cell proliferation, motility and their participation in tumour progression using in-vivo models. Tumour microenvironment is currently the focus of many researches and the highlighting of the relationship between cancer cells and surrounding elements, is expanding. One of the major physic-chemical parameter involved in tumour progression is the hypoxia conditions observed in solid cancer. Due to their position on the cell membrane, ion channels are good candidates to transduce or to be modulated by environmental modifications. Until now, few reports have been interested in the modification of ion channel activities or expression in this context, compared to other pathological situations such as ischemia reperfusion. The aim of our review is to summarize the current knowledge about the calcium and potassium channels properties in the context of hypoxia in tumours. This review could pave the way to orientate new studies around this exciting field to obtain new potential therapeutic approaches.


Asunto(s)
Canales de Calcio/metabolismo , Neoplasias/metabolismo , Oxígeno/metabolismo , Canales de Potasio/metabolismo , Microambiente Tumoral , Animales , Hipoxia de la Célula , Humanos
14.
Front Pharmacol ; 11: 568993, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33178018

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is the fourth most common cause of cancer-related deaths in United States and Europe. It is predicted that PDAC will become the second leading cause of cancer-related deaths during the next decades. The development of PDAC is not well understood, however, studies have shown that dysregulated exocrine pancreatic fluid secretion can contribute to pathologies of exocrine pancreas, including PDAC. The major roles of healthy exocrine pancreatic tissue are secretion of enzymes and bicarbonate rich fluid, where ion channels participate to fine-tune these biological processes. It is well known that ion channels located in the plasma membrane regulate multiple cellular functions and are involved in the communication between extracellular events and intracellular signaling pathways and can function as signal transducers themselves. Hereby, they contribute to maintain resting membrane potential, electrical signaling in excitable cells, and ion homeostasis. Despite their contribution to basic cellular processes, ion channels are also involved in the malignant transformation from a normal to a malignant phenotype. Aberrant expression and activity of ion channels have an impact on essentially all hallmarks of cancer defined as; uncontrolled proliferation, evasion of apoptosis, sustained angiogenesis and promotion of invasion and migration. Research indicates that certain ion channels are involved in the aberrant tumor growth and metastatic processes of PDAC. The purpose of this review is to summarize the important expression, localization, and function of ion channels in normal exocrine pancreatic tissue and how they are involved in PDAC progression and development. As ion channels are suggested to be potential targets of treatment they are furthermore suggested to be biomarkers of different cancers. Therefore, we describe the importance of ion channels in PDAC as markers of diagnosis and clinical factors.

15.
Cell Physiol Biochem ; 23(4-6): 335-46, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19471101

RESUMEN

The calcium-sensing receptor (CaR), is a G protein-dependent receptor that responds to increments in extracellular Ca(2+) ([Ca(2+)](o)). We previously reported that an increase in [Ca(2+)](o) induced a release of intracellular calcium and Ca(2+) entry via store operated channels (SOCs). We also demonstrated that MCF-7 cells express Transient Receptor Potential canonical 1 (TRPC1) channels. Herein, we investigated CaR intracellular signaling pathways and examined the role of TRPC1 in CaR-induced cell proliferation, through the extracellular signal-regulated Kinases 1 & 2 (ERK1/2) pathways. Treatment by [Ca(2+)](o) increased both MCF-7 cell proliferation and TRPC1 expression. Both the [Ca(2+)](o) proliferative effect and TRPC1 protein levels were abolished by the ERK1/2 inhibitors. Moreover, [Ca(2+)](o) failed to increase cell proliferation either in the presence of CaR or TRPC1 siRNAs. Both [Ca(2+)](o) and the selective CaR activator spermine, elicited time and dose-dependent ERK1/2 phosphorylation. ERK1/2 phosphorylation was almost completely inhibited by treatment with the phospholipase C and the protein kinase C inhibitors. Treatment with 2-aminoethoxydiphenyl borate (2-APB), and SKF-96365 or by siTRPC1 diminished both [Ca(2+)](o)- and spermine-stimulated ERK1/2 phosphorylation. Moreover, down-regulation of TRPC1 by siRNA reduced the Ca(2+) entry induced by CaR activation. We conclude that the CaR activates ERK1/2 via a PLC/PKC-dependent pathway. Moreover, TRPC1 is required for the ERK1/2 phosphorylation, Ca(2+) entry and the CaR-proliferative effect.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Receptores Sensibles al Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/patología , Calcio/metabolismo , Proliferación Celular , Regulación hacia Abajo , Femenino , Humanos , Fosforilación , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Células Tumorales Cultivadas , Fosfolipasas de Tipo C/metabolismo
16.
Arch Biochem Biophys ; 486(1): 58-63, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19332022

RESUMEN

The calcium sensing receptor (CaR) is a G-protein-coupled receptor that is activated by extracellular calcium ([Ca(2+)](o)). In MCF-7 human breast cancer cells, we previously reported that treatment with [Ca(2+)](o) for 24h leads to an over-expression of the Transient Receptor Potential Canonical 1 (TRPC1) cation channel and cell proliferation. Both involve the extracellular signal-regulated Kinases 1 & 2 (ERK1/2). MCF-7 also expressed epidermal growth factor receptor (EGFR) which is involved in cell proliferation through ERK1/2. Therefore, we investigated the cross-talk between CaR and EGFR in mediating ERK1/2 phosphorylation, TRPC1 over-expression and cell proliferation. Our data show that both high [Ca(2+)](o) and EGF phosphorylate ERK1/2. Furthermore, inhibition of EGFR kinase and matrix metalloproteinases (MMPs) reduced the overall effects mediated by [Ca(2+)](o) such as activation of ERK1/2, expression of TRPC1 and cell proliferation. They indicate the important role of the CaR-EGFR-ERK axis in transmitting mitogenic signals generated by high [Ca(2+)](o) in MCF-7 cells.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Calcio/farmacología , Receptores ErbB/metabolismo , Receptores Sensibles al Calcio/metabolismo , Canales Catiónicos TRPC/metabolismo , Secuencia de Bases , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cartilla de ADN/genética , Dipéptidos/farmacología , Activación Enzimática/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Quinazolinas , Canales Catiónicos TRPC/genética , Activación Transcripcional/efectos de los fármacos , Tirfostinos/farmacología
17.
BMC Cancer ; 8: 125, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18452628

RESUMEN

BACKGROUND: TRP channels have been shown to be involved in tumour generation and malignant growth. However, the expression of these channels in breast cancer remains unclear. Here we studied the expression and function of endogenous TRPC6 channels in a breast cancer cell line (MCF-7), a human breast cancer epithelial primary culture (hBCE) and in normal and tumour breast tissues. METHODS: Molecular (Western blot and RT-PCR), and immunohistochemical techniques were used to investigate TRPC6 expression. To investigate the channel activity in both MCF-7 cells and hBCE we used electrophysiological technique (whole cell patch clamp configuration). RESULTS: A non selective cationic current was activated by the oleoyl-2-acetyl-sn-glycerol (OAG) in both hBCE and MCF-7 cells. OAG-inward current was inhibited by 2-APB, SK&F 96365 and La3+. TRPC6, but not TRPM7, was expressed both in hBCE and in MCF-7 cells. TRPC3 was only expressed in hBCE. Clinically, TRPC6 mRNA and protein were elevated in breast carcinoma specimens in comparison to normal breast tissue. Furthermore, we found that the overexpression of TRPC6 protein levels were not correlated with tumour grades, estrogen receptor expression or lymph node positive tumours. CONCLUSION: Our results indicate that TRPC6 channels are strongly expressed and functional in breast cancer epithelial cells. Moreover, the overexpression of these channels appears without any correlation with tumour grade, ER expression and lymph node metastasis. Our findings support the idea that TRPC6 may have a role in breast carcinogenesis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Glándulas Mamarias Humanas/metabolismo , Neoplasias Glandulares y Epiteliales/metabolismo , Canales Catiónicos TRPC/biosíntesis , Western Blotting , Neoplasias de la Mama/patología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Glándulas Mamarias Humanas/citología , Estadificación de Neoplasias , Neoplasias Glandulares y Epiteliales/patología , Técnicas de Placa-Clamp , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPC/fisiología , Canal Catiónico TRPC6
18.
Oncotarget ; 7(49): 81588-81597, 2016 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-27835593

RESUMEN

Orai3 channel has emerged as important player in malignant transformation. Indeed, its expression is increased in cancer and favors cell proliferation and survival by permitting calcium influx. In this study, Orai3 was overexpressed in lung adenocarcinoma as compared to their matched non-tumour samples and was associated with tumoural aggressiveness. Moreover, its expression was associated with estrogen receptor alpha (ERα) expression and visceral pleural invasion in multivariate analysis. Furthermore, both the overall survival (OS) median and the metastasis free survival (MFS) median of tumors with high Orai3 expression were lower than in low Orai3 expression regardless of cancer stage (35.01 months vs. 51.11 months for OS and 46.01 months vs. 62.04 months for MFS). In conclusion, Orai3 protein level constitutes an independent prognostic marker in lung adenocarcinoma, and a novel prognostic marker that could help selecting the patients with worst prognosis to be treated with adjuvant chemotherapy in resectable stage.


Asunto(s)
Adenocarcinoma/química , Adenocarcinoma/cirugía , Biomarcadores de Tumor/análisis , Neoplasias Pulmonares/química , Neoplasias Pulmonares/cirugía , Proteína ORAI1/análisis , Neumonectomía , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adenocarcinoma del Pulmón , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia sin Enfermedad , Receptor alfa de Estrógeno/análisis , Femenino , Humanos , Modelos Logísticos , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Análisis Multivariante , Invasividad Neoplásica , Estadificación de Neoplasias , Oportunidad Relativa , Neumonectomía/efectos adversos , Neumonectomía/mortalidad , Modelos de Riesgos Proporcionales , Factores de Riesgo , Factores de Tiempo , Resultado del Tratamiento , Regulación hacia Arriba
19.
Oncotarget ; 7(24): 36419-36435, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27183905

RESUMEN

Intracellular Ca2+ levels are important regulators of cell cycle and proliferation. We, and others, have previously reported the role of KCa3.1 (KCNN4) channels in regulating the membrane potential and the Ca2+ entry in association with cell proliferation. However, the relevance of KC3.1 channels in cancer prognosis as well as the molecular mechanism of Ca2+ entry triggered by their activation remain undetermined. Here, we show that RNAi-mediated knockdown of KCa3.1 and/or TRPC1 leads to a significant decrease in cell proliferation due to cell cycle arrest in the G1 phase. These results are consistent with the observed upregulation of both channels in synchronized cells at the end of G1 phase. Additionally, knockdown of TRPC1 suppressed the Ca2+ entry induced by 1-EBIO-mediated KCa3.1 activation, suggesting a functional cooperation between TRPC1 and KCa3.1 in the regulation of Ca2+ entry, possibly within lipid raft microdomains where these two channels seem to co-localize. We also show significant correlations between KCa3.1 mRNA expression and poor patient prognosis and unfavorable clinical breast cancer parameters by mining large datasets in the public domain. Together, these results highlight the importance of KCa3.1 in regulating the proliferative mechanisms in breast cancer cells as well as in providing a promising novel target in prognosis and therapy.


Asunto(s)
Neoplasias de la Mama/genética , Proliferación Celular/genética , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/genética , Canales Catiónicos TRPC/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Calcio/metabolismo , Femenino , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Canales de Potasio de Conductancia Intermedia Activados por el Calcio/metabolismo , Estimación de Kaplan-Meier , Células MCF-7 , Pronóstico , Interferencia de ARN , Canales Catiónicos TRPC/metabolismo
20.
Front Physiol ; 4: 220, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23970866

RESUMEN

K(+) ions play a major role in many cellular processes. The deregulation of K(+) signaling is associated with a variety of diseases such as hypertension, atherosclerosis, or diabetes. K(+) ions are important for setting the membrane potential, the driving force for Ca(2+) influx, and regulate volume of growing cells. Moreover, it is increasingly recognized that K(+) channels control cell proliferation through a novel signaling mechanisms triggered and modulated independently of ion fluxes. In cancer, aberrant expression, regulation and/or sublocalization of K(+) channels can alter the downstream signals that converge on the cell cycle machinery. Various K(+) channels are involved in cell cycle progression and are needed only at particular stages of the cell cycle. Consistent with this idea, the expression of Eag1 and HERG channels fluctuate along the cell cycle. Despite of acquired knowledge, our understanding of K(+) channels functioning in cancer cells requires further studies. These include identifying the molecular mechanisms controlling the cell cycle machinery. By understanding how K(+) channels regulate cell cycle progression in cancer cells, we will gain insights into how cancer cells subvert the need for K(+) signal and its downstream targets to proliferate.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA