Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell Calcium ; 113: 102760, 2023 07.
Article in English | MEDLINE | ID: mdl-37247443

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Female , Humans , Breast Neoplasms/metabolism , Calcium
2.
Cancers (Basel) ; 14(19)2022 Oct 09.
Article in English | MEDLINE | ID: mdl-36230869

ABSTRACT

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.

3.
Int J Mol Sci ; 23(18)2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36142596

ABSTRACT

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.


Subject(s)
Cisplatin , Neoplasms , Calcium/metabolism , Cell Line, Tumor , Cisplatin/metabolism , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism
4.
Int J Mol Sci ; 23(14)2022 Jul 18.
Article in English | MEDLINE | ID: mdl-35887266

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Calcium/metabolism , Calmodulin/metabolism , Carcinoma, Pancreatic Ductal/genetics , Cell Proliferation , Humans , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , TRPC Cation Channels/genetics , TRPC Cation Channels/metabolism , Pancreatic Neoplasms
5.
Article in English | MEDLINE | ID: mdl-32761457

ABSTRACT

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.


Subject(s)
Breast Neoplasms , Female , Humans , Ion Channels , Signal Transduction , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
6.
Front Pharmacol ; 11: 568993, 2020.
Article in English | MEDLINE | ID: mdl-33178018

ABSTRACT

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.

7.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118644, 2020 04.
Article in English | MEDLINE | ID: mdl-31931022

ABSTRACT

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.


Subject(s)
Calcium Channels/metabolism , Neoplasms/metabolism , Oxygen/metabolism , Potassium Channels/metabolism , Tumor Microenvironment , Animals , Cell Hypoxia , Humans
8.
J Cell Physiol ; 232(12): 3631-3640, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28145580

ABSTRACT

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.


Subject(s)
Breast Neoplasms/drug therapy , Cell Movement/drug effects , Focal Adhesion Kinase 1/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Indolizines/pharmacokinetics , Integrin beta1/metabolism , Neoplasm Proteins/metabolism , Stromal Interaction Molecule 1/metabolism , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glycosylation , Humans , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Protein Processing, Post-Translational/drug effects , RNA Interference , Signal Transduction/drug effects , Stromal Interaction Molecule 1/genetics , Time Factors , Transfection
9.
Oncotarget ; 7(49): 81588-81597, 2016 Dec 06.
Article in English | MEDLINE | ID: mdl-27835593

ABSTRACT

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.


Subject(s)
Adenocarcinoma/chemistry , Adenocarcinoma/surgery , Biomarkers, Tumor/analysis , Lung Neoplasms/chemistry , Lung Neoplasms/surgery , ORAI1 Protein/analysis , Pneumonectomy , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Estrogen Receptor alpha/analysis , Female , Humans , Logistic Models , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Staging , Odds Ratio , Pneumonectomy/adverse effects , Pneumonectomy/mortality , Proportional Hazards Models , Risk Factors , Time Factors , Treatment Outcome , Up-Regulation
10.
Oncotarget ; 7(24): 36419-36435, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27183905

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Cell Proliferation/genetics , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , TRPC Cation Channels/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/metabolism , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Kaplan-Meier Estimate , MCF-7 Cells , Prognosis , RNA Interference , TRPC Cation Channels/metabolism
11.
Pflugers Arch ; 468(5): 751-62, 2016 05.
Article in English | MEDLINE | ID: mdl-26743871

ABSTRACT

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.


Subject(s)
Carcinogenesis/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Tumor Microenvironment , Animals , Cell Movement , Cell Proliferation , Ether-A-Go-Go Potassium Channels/genetics , Humans , Neurons/metabolism , Neurons/physiology
12.
Biochim Biophys Acta ; 1848(10 Pt B): 2621-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25703813

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/genetics , DNA, Neoplasm/metabolism , Epigenesis, Genetic , Neoplasms/diagnosis , Neoplasms/genetics , Biomarkers, Tumor/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , DNA Methylation , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/metabolism , Neoplasms/pathology , Potassium Channels/genetics , Potassium Channels/metabolism , Promoter Regions, Genetic , Receptors, Ionotropic Glutamate/genetics , Receptors, Ionotropic Glutamate/metabolism , Signal Transduction , Tumor Cells, Cultured
13.
PLoS One ; 8(10): e76411, 2013.
Article in English | MEDLINE | ID: mdl-24124558

ABSTRACT

sp²-Iminosugar-type castanospermine analogues have been shown to exhibit anti-tumor activity. However, their effects on cell proliferation and apoptosis and the molecular mechanism at play are not fully understood. Here, we investigated the effect of two representatives, namely the pseudo-S- and C-octyl glycoside 2-oxa-3-oxocastanospermine derivatives SO-OCS and CO-OCS, on MCF-7 and MDA-MB-231 breast cancer and MCF-10A mammary normal cell lines. We found that SO-OCS and CO-OCS inhibited breast cancer cell viability in a concentration- and time-dependent manner. This effect is specific to breast cancer cells as both molecules had no impact on normal MCF-10A cell proliferation. Both drugs induced a cell cycle arrest. CO-OCS arrested cell cycle at G1 and G2/M in MCF-7 and MDA-MB-231 cells respectively. In MCF-7 cells, the G1 arrest is associated with a reduction of CDK4 (cyclin-dependent kinase 4), cyclin D1 and cyclin E expression, pRb phosphorylation, and an overexpression of p21(Waf1/Cip1). In MDA-MB-231 cells, CO-OCS reduced CDK1 but not cyclin B1 expression. SO-OCS accumulated cells in G2/M in both cell lines and this blockade was accompanied by a decrease of CDK1, but not cyclin B1 expression. Furthermore, both drugs induced apoptosis as demonstrated by the increased percentage of annexin V positive cells and Bax/Bcl-2 ratio. Interestingly, in normal MCF-10A cells the two drugs failed to modify cell proliferation, cell cycle progression, cyclins, or CDKs expression. These results demonstrate that the effect of CO-OCS and SO-OCS is triggered by both cell cycle arrest and apoptosis, suggesting that these castanospermine analogues may constitute potential anti-cancer agents against breast cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Indolizines/pharmacology , Breast Neoplasms/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans
14.
PLoS One ; 8(9): e72889, 2013.
Article in English | MEDLINE | ID: mdl-24058448

ABSTRACT

Orai channels have been associated with cell proliferation, survival and metastasis in several cancers. The present study investigates the expression and the role of Orai3 in cell proliferation in non-small cell lung cancer (NSCLC). We show that Orai3 is over-expressed in cancer tissues as compared to the non-tumoral ones. Furthermore, Orai3 staining is stronger in high grade tumors. Pharmacological inhibition or knockdown of Orai3 significantly reduced store operated calcium entry (SOCE), inhibited cell proliferation and arrested cells of two NSCLC cell lines in G0/G1 phase. These effects were concomitant with a down-regulation of cyclin D1, cyclin E, CDK4 and CDK2 expression. Moreover, Orai3 silencing decreased Akt phosphorylation levels. In conclusion, Orai3 constitutes a native SOCE pathway in NSCLC that controls cell proliferation and cell cycle progression likely via Akt pathway.


Subject(s)
Adenocarcinoma/genetics , Calcium Channels/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-akt/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Calcium/metabolism , Calcium Channels/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin E/genetics , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Female , G1 Phase Cell Cycle Checkpoints , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Resting Phase, Cell Cycle , Signal Transduction
15.
Front Physiol ; 4: 220, 2013.
Article in English | MEDLINE | ID: mdl-23970866

ABSTRACT

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.

16.
Trends Mol Med ; 19(2): 117-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23253476

ABSTRACT

Breast cancer is the most frequently occurring cancer in women and has the highest rate of mortality. Ion channels such as the transient receptor potential (TRP) channels could play a critical role in the development and progression of cancer. Although these channels are frequently and abundantly expressed in many tumors, their expression, activity, and roles in the context of breast cancer remain poorly understood. This review summarizes our current knowledge regarding TRP channels expressed in human breast tissue, primary human breast epithelial cells, and cell lines, the functional role of TRP channels during breast cancer cell growth and migration, as well as their relationship with clinical and pathological features.


Subject(s)
Breast Neoplasms/metabolism , Transient Receptor Potential Channels/metabolism , Animals , Biomarkers, Tumor , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Epithelial Cells/metabolism , Female , Gene Expression Regulation , Humans , Mammary Glands, Animal/metabolism , Mammary Glands, Human/metabolism , Molecular Targeted Therapy , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
17.
Biochim Biophys Acta ; 1833(3): 752-60, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23266555

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Calcium Channels/metabolism , Cell Proliferation , G1 Phase/physiology , Proto-Oncogene Proteins c-myc/metabolism , Adenocarcinoma , Apoptosis , Blotting, Western , Breast/cytology , Breast/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Cells, Cultured , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Immunoenzyme Techniques , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tissue Array Analysis
18.
Bull Cancer ; 99(6): 655-64, 2012 Jun.
Article in French | MEDLINE | ID: mdl-22640890

ABSTRACT

Breast cancer (BC) has the highest incidence rate in women in industrialized countries. Statistically, it is estimated that one out of 10 women will develop BC during her life. Evidence is accumulating for the role of ion channels in the development of cancer. Most studied ion channels in BC are K(+) channels, which are involved in cell proliferation, cell cycle progression and cell migration, and Na(+) channels, which correlate with invasiveness. Emerging studies demonstrated the role of Ca(2+) signaling in cancer cell proliferation, survival and migration. Recent findings demonstrated that the expression and/or activity of the transient receptor potential (TRP) channels are altered in several cancers. Among the TRP families, TRPC (canonical or classical), TRPM (melastatin) and TRPV (vanilloid) are related to malignant growth and cancer progression. Although these channels are frequently and abundantly expressed in many tumors, their specific expression, activity and roles in BC are still poorly understood. The expression of TRP channels has also been proposed as a tool for diagnosis, prognosis and/or therapeutic issues of several diseases. In cancer, TRPV6 and TRPM8 have been proposed as tumor progression markers of prostate cancer outcome and TRPC6 as a novel therapeutic target for esophageal carcinoma. Interestingly high levels of TRPC3 expression correlate with a favorable prognosis in patients with lung adenocarcinoma. Our team has recently reported the expression and role of TRPC1, TRPC6, TRPM7, TRPM8 and TRPV6 in BC cell lines and primary cultures. We have also investigated TRP expression and their clinical significance in human breast adenocarcinoma and we suggest that TRP channels are new potential BC markers. Indeed TRPC1 and TRPM8 may be considered as good prognosis markers of well-differentiated tumors, TRPM7 as a proliferative marker of poorly differentiated tumors and TRPV6 as a prognosis marker of aggressive cancers. In this review, we summarize the data reported to date regarding the changes in TRP expression associated with BC. We also discuss the importance of TRP channels in BC cells proliferation and migration and their interest as new BC markers.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Transient Receptor Potential Channels/physiology , Biomarkers, Tumor/physiology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Female , Humans , Prognosis
19.
Cell Physiol Biochem ; 28(5): 813-22, 2011.
Article in English | MEDLINE | ID: mdl-22178934

ABSTRACT

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.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Transient Receptor Potential Channels/metabolism , Breast Neoplasms/metabolism , Cells, Cultured , Female , Humans , Immunohistochemistry , Neoplasm Staging , Protein Serine-Threonine Kinases , RNA Interference , RNA, Small Interfering/metabolism , TRPC Cation Channels/metabolism , TRPC6 Cation Channel , TRPM Cation Channels/metabolism , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/genetics
20.
J Cell Physiol ; 226(2): 542-51, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20683915

ABSTRACT

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.


Subject(s)
Apoptosis/physiology , Breast Neoplasms , Breast/cytology , Calcium Channels/metabolism , Cell Cycle/physiology , Epithelial Cells/physiology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium Channels/genetics , Cell Proliferation , Cell Survival , Cells, Cultured , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , Down-Regulation , Epithelial Cells/cytology , Female , Humans , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL