Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-32761457

RESUMO

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.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Canais Iônicos , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Microambiente Tumoral
2.
Int J Mol Sci ; 23(18)2022 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-36142596

RESUMO

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.


Assuntos
Cisplatino , Neoplasias , Cálcio/metabolismo , Linhagem Celular Tumoral , Cisplatino/metabolismo , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Humanos , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo
3.
Int J Mol Sci ; 23(6)2022 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-35328381

RESUMO

Breast cancer is the leading cause of cancer death among women in worldwide and France. The disease prognosis and treatment differ from one breast cancer subtype to another, and the disease outcome depends on many prognostic factors. Deregulation of ion flux (especially Ca2+ flux) is involved in many pathophysiology processes, including carcinogenesis. Inside the cell, the inositol-trisphosphate receptor (IP3R) is a major player in the regulation of the Ca2+ flux from the endoplasmic reticulum to the cytoplasm. The IP3Rs (and particularly the IP3R3 subtype) are known to be involved in proliferation, migration, and invasion processes in breast cancer cell lines. The objective of the present study was to evaluate the potential value of IP3Rs as prognostic biomarkers in breast cancer. We found that expression levels of IP3R3 and IP3R1 (but not IP3R2) were significantly higher in invasive breast cancer of no special type than in non-tumor tissue from the same patient. However, the IP3R3 subtype was expressed more strongly than the IP3R1 and IP3R2 subtypes. Furthermore, the expression of IP3R3 (but not of IP3R1 or IP3R2) was positively correlated with prognostic factors such as tumor size, regional node invasion, histologic grade, proliferation index, and hormone receptor status. In an analysis of public databases, we found that all IP3Rs types are significantly associated with overall survival and progression-free survival in patients with breast cancer. We conclude that relative to the other two IP3R subtypes, IP3R3 expression is upregulated in breast cancer and is correlated with prognostic factors.


Assuntos
Neoplasias da Mama , Neoplasias da Mama/metabolismo , Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Feminino , Humanos , Inositol/metabolismo , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Prognóstico
4.
Int J Mol Sci ; 23(14)2022 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-35887266

RESUMO

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.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Cálcio/metabolismo , Calmodulina/metabolismo , Carcinoma Ductal Pancreático/genética , Proliferação de Células , Humanos , Neoplasias Pancreáticas/genética , Fosfatidilinositol 3-Quinases/metabolismo , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Neoplasias Pancreáticas
5.
Haematologica ; 105(3): 610-622, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31413092

RESUMO

Hereditary xerocytosis is a dominantly inherited red cell membrane disorder caused in most cases by gain-of-function mutations in PIEZO1, encoding a mechanosensitive ion channel that translates a mechanic stimulus into calcium influx. We found that PIEZO1 was expressed early in erythroid progenitor cells, and investigated whether it could be involved in erythropoiesis, besides having a role in the homeostasis of mature red cell hydration. In UT7 cells, chemical PIEZO1 activation using YODA1 repressed glycophorin A expression by 75%. This effect was PIEZO1-dependent since it was reverted using specific short hairpin-RNA knockdown. The effect of PIEZO1 activation was confirmed in human primary progenitor cells, maintaining cells at an immature stage for longer and modifying the transcriptional balance in favor of genes associated with early erythropoiesis, as shown by a high GATA2/GATA1 ratio and decreased α/ß-globin expression. The cell proliferation rate was also reduced, with accumulation of cells in G0/G1 of the cell cycle. The PIEZO1-mediated effect on UT7 cells required calcium-dependent activation of the NFAT and ERK1/2 pathways. In primary erythroid cells, PIEZO1 activation synergized with erythropoietin to activate STAT5 and ERK, indicating that it may modulate signaling pathways downstream of erythropoietin receptor activation. Finally, we studied the in-vitro erythroid differentiation of primary cells obtained from 14 PIEZO1-mutated patients, from 11 families, carrying ten different mutations. We observed a delay in erythroid differentiation in all cases, ranging from mild (n=3) to marked (n=8). Overall, these data demonstrate a role for PIEZO1 during erythropoiesis, since activation of PIEZO1 - both chemically and through activating mutations - delays erythroid maturation, providing new insights into the pathophysiology of hereditary xerocytosis.


Assuntos
Anemia Hemolítica Congênita , Canais Iônicos , Anemia Hemolítica Congênita/genética , Diferenciação Celular , Eritropoese/genética , Humanos , Hidropisia Fetal , Canais Iônicos/genética , Células-Tronco
6.
Arch Toxicol ; 94(3): 735-747, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32080757

RESUMO

Cadmium is a xenobiotic involved in neoplastic transformation. Cadmium enters the cells through divalent cation transporters including the Transient Receptor Potential Melastatin-related 7 (TRPM7) which is known to be involved in cancer cell fate. This work aimed to study the role of TRPM7 in neoplastic transformation induced by cadmium exposure in non-cancer epithelial cells. Non-cancer epithelial cells were chronically exposed to low-dose of cadmium. TRPM7 expression and function were studied by Western-Blot, Patch-Clamp and calcium and magnesium imaging. Finally, cell migration and invasion were studied by Boyden chamber assays. Chronic cadmium exposure induced TRPM7 overexpression and increased the membrane currents (P < 0.001). Cells exposed to cadmium had higher intracellular calcium and magnesium levels (P < 0.05). TRPM7 silencing restored calcium levels but strongly decreased intracellular magnesium concentration (P < 0.001). Moreover, cadmium exposure enhanced both cell migration and invasion, but TRPM7 silencing strongly decreased these features (P < 0.001). Furthermore, mammary epithelial cells exposed to cadmium became rounded and had less cell-to-cell junctions. Cadmium exposure decreased epithelial markers while the mesenchymal ones were increased. Importantly, TRPM7 silencing was able to reverse these phenotypic modifications (P < 0.05). To summarize, our data show that chronic cadmium exposure enhanced TRPM7 expression and activity in non-cancer epithelial cells. TRPM7 overexpression induced intracellular magnesium increase and stimulated cell migration and invasion. These neoplastic properties could be linked to a TRPM7-dependent epithelial-to-mesenchymal transition reprogramming in cell exposed to cadmium. These findings provide new insights into the regulation of cell fates by cadmium exposure.


Assuntos
Cádmio/toxicidade , Movimento Celular/efeitos dos fármacos , Substâncias Perigosas/toxicidade , Canais de Cátion TRPM/metabolismo , Transição Epitelial-Mesenquimal , Humanos
7.
Biochim Biophys Acta Mol Cell Res ; 1865(7): 945-958, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29630900

RESUMO

Cell morphology is altered in the migration process, and the underlying cytoskeleton remodeling is highly dependent of intracellular Ca2+ concentration. Many calcium channels are known to be involved in migration. Inositol 1,4,5-trisphosphate receptor (IP3R) was demonstrated to be implicated in breast cancer cells migration, but its involvement in morphological changes during the migration process remains unclear. In the present work, we showed that IP3R3 expression was correlated to cell morphology. IP3R3 silencing induced rounding shape and decreased adhesion in invasive breast cancer cell lines. Moreover, IP3R3 silencing decreased ARHGAP18 expression, RhoA activity, Cdc42 expression and Y861FAK phosphorylation. Interestingly, IP3R3 was able to regulate profilin remodeling, without inducing any myosin II reorganization. IP3R3 silencing revealed an oscillatory calcium signature, with a predominant oscillating profile occurring in early wound repair. To summarize, we demonstrated that IP3R3 is able to modulate intracellular Ca2+ availability and to coordinate the remodeling of profilin cytoskeleton organization through the ARHGAP18/RhoA/mDia1/FAK pathway.


Assuntos
Neoplasias da Mama/genética , Receptores de Inositol 1,4,5-Trifosfato/genética , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Profilinas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/metabolismo , Cálcio/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Feminino , Quinase 1 de Adesão Focal/metabolismo , Forminas , Proteínas Ativadoras de GTPase/metabolismo , Inativação Gênica , Humanos , Células MCF-7 , Fosforilação , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Br J Cancer ; 120(4): 453-465, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30739912

RESUMO

BACKGROUND: Carcinogenesis occurs in elastin-rich tissues and leads to local inflammation and elastolytic proteinase release. This contributes to bioactive matrix fragment (Matrikine) accumulation like elastin degradation products (EDP) stimulating tumour cell invasive and metastatic properties. We previously demonstrate that EDPs exert protumoural activities through Hsp90 secretion to stabilised extracellular proteinases. METHODS: EDP influence on cancer cell blebbing and extracellular vesicle shedding were examined with a videomicroscope coupled with confocal Yokogawa spinning disk, by transmission electron microscopy, scanning electron microscopy and confocal microscopy. The ribosomal protein SA (RPSA) elastin receptor was identified after affinity chromatography by western blotting and cell immunolocalisation. mRNA expression was studied using real-time PCR. SiRNA were used to confirm the essential role of RPSA. RESULTS: We demonstrate that extracellular matrix degradation products like EDPs induce tumour amoeboid phenotype with cell membrane blebbing and shedding of extracellular vesicle containing Hsp90 and proteinases in the extracellular space. EDPs influence intracellular calcium influx and cytoskeleton reorganisation. Among matrikines, VGVAPG and AGVPGLGVG peptides reproduced EDP effects through RPSA binding. CONCLUSIONS: Our data suggests that matrikines induce cancer cell blebbing and extracellular vesicle release through RPSA binding, favouring dissemination, cell-to-cell communication and growth of cancer cells in metastatic sites.


Assuntos
Proteínas da Matriz Extracelular/farmacologia , Vesículas Extracelulares/fisiologia , Neoplasias/patologia , Fragmentos de Peptídeos/farmacologia , Receptores de Laminina/metabolismo , Proteínas Ribossômicas/metabolismo , Amidas/farmacologia , Cálcio/metabolismo , Comunicação Celular , Linhagem Celular Tumoral , Elastina/farmacologia , Proteínas de Choque Térmico HSP90/análise , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Neoplasias/metabolismo , Piridinas/farmacologia , Transdução de Sinais , Quinases Associadas a rho/fisiologia
9.
J Cell Physiol ; 232(12): 3631-3640, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28145580

RESUMO

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.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Movimento Celular/efeitos dos fármacos , Quinase 1 de Adesão Focal/metabolismo , Inibidores de Glicosídeo Hidrolases/farmacologia , Indolizinas/farmacocinética , Integrina beta1/metabolismo , Proteínas de Neoplasias/metabolismo , Molécula 1 de Interação Estromal/metabolismo , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Ativação Enzimática , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Glicosilação , Humanos , Células MCF-7 , Invasividade Neoplásica , Proteínas de Neoplasias/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Molécula 1 de Interação Estromal/genética , Fatores de Tempo , Transfecção
10.
Biochim Biophys Acta ; 1848(10 Pt B): 2621-8, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25703813

RESUMO

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.


Assuntos
Biomarcadores Tumorais/genética , DNA de Neoplasias/metabolismo , Epigênese Genética , Neoplasias/diagnóstico , Neoplasias/genética , Biomarcadores Tumorais/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Metilação de DNA , DNA de Neoplasias/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Canais de Potássio/genética , Canais de Potássio/metabolismo , Regiões Promotoras Genéticas , Receptores Ionotrópicos de Glutamato/genética , Receptores Ionotrópicos de Glutamato/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas
11.
Pflugers Arch ; 468(5): 751-62, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26743871

RESUMO

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.


Assuntos
Carcinogênese/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Microambiente Tumoral , Animais , Movimento Celular , Proliferação de Células , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Neurônios/metabolismo , Neurônios/fisiologia
12.
Int J Cancer ; 137(6): 1306-17, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25704182

RESUMO

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


Assuntos
Epigênese Genética/genética , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Neoplasias Pulmonares/genética , Adenocarcinoma/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Metilação de DNA/genética , Epigenômica/métodos , Feminino , Xenoenxertos , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Prognóstico , Regiões Promotoras Genéticas/genética
13.
Hum Mutat ; 35(10): 1221-32, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25044882

RESUMO

Stormorken syndrome is a rare autosomal dominant disorder characterized by a phenotype that includes miosis, thrombocytopenia/thrombocytopathy with bleeding time diathesis, intellectual disability, mild hypocalcemia, muscle fatigue, asplenia, and ichthyosis. Using targeted sequencing and whole-exome sequencing, we identified the c.910C > T transition in a STIM1 allele (p.R304W) only in patients and not in their unaffected family members. STIM1 encodes stromal interaction molecule 1 protein (STIM1), which is a finely tuned endoplasmic reticulum Ca(2+) sensor. The effect of the mutation on the structure of STIM1 was investigated by molecular modeling, and its effect on function was explored by calcium imaging experiments. Results obtained from calcium imaging experiments using transfected cells together with fibroblasts from one patient are in agreement with impairment of calcium homeostasis. We show that the STIM1 p.R304W variant may affect the conformation of the inhibitory helix and unlock the inhibitory state of STIM1. The p.R304W mutation causes a gain of function effect associated with an increase in both resting Ca(2+) levels and store-operated calcium entry. Our study provides evidence that Stormorken syndrome may result from a single-gene defect, which is consistent with Mendelian-dominant inheritance.


Assuntos
Transtornos Plaquetários/genética , Dislexia/genética , Ictiose/genética , Proteínas de Membrana/genética , Transtornos de Enxaqueca/genética , Miose/genética , Proteínas de Neoplasias/genética , Mutação Puntual , Baço/anormalidades , Adolescente , Adulto , Idoso , Transtornos Plaquetários/metabolismo , Transtornos Plaquetários/patologia , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Criança , Pré-Escolar , Dislexia/metabolismo , Dislexia/patologia , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/ultraestrutura , Eritrócitos Anormais/metabolismo , Eritrócitos Anormais/patologia , Feminino , Humanos , Ictiose/metabolismo , Ictiose/patologia , Lactente , Recém-Nascido , Masculino , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Pessoa de Meia-Idade , Transtornos de Enxaqueca/metabolismo , Transtornos de Enxaqueca/patologia , Miose/metabolismo , Miose/patologia , Fadiga Muscular/genética , Fibras Musculares Esqueléticas/patologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Linhagem , Estrutura Secundária de Proteína , Baço/metabolismo , Baço/patologia , Molécula 1 de Interação Estromal
14.
Biochim Biophys Acta ; 1833(3): 752-60, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23266555

RESUMO

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.


Assuntos
Neoplasias da Mama/patologia , Canais de Cálcio/metabolismo , Proliferação de Células , Fase G1/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , Adenocarcinoma , Apoptose , Western Blotting , Mama/citologia , Mama/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Cálcio/metabolismo , Canais de Cálcio/química , Canais de Cálcio/genética , Células Cultivadas , Feminino , Imunofluorescência , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Técnicas Imunoenzimáticas , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-myc/genética , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Análise Serial de Tecidos
15.
Cell Calcium ; 113: 102760, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37247443

RESUMO

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.


Assuntos
Neoplasias da Mama , Feminino , Humanos , Neoplasias da Mama/metabolismo , Cálcio
16.
Biochim Biophys Acta ; 1813(5): 723-30, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21315112

RESUMO

Insulin-like Growth Factor-1 (IGF-1) plays a key role in breast cancer development and cell cycle regulation. It has been demonstrated that IGF-1 stimulates cyclin expression, thus regulating the G1 to S phase transition of the cell cycle. Potassium (K(+)) channels are involved in the G1 phase progression of the cell cycle induced by growth factors. However, mechanisms that allow growth factors to cooperate with K(+) channels in order to modulate the G1 phase progression and cyclin expression remain unknown. Here, we focused on hEag1 K(+) channels which are over-expressed in breast cancer and are involved in the G1 phase progression of breast cancer cells (MCF-7). As expected, IGF-1 increased cyclin D1 and E expression of MCF-7 cells in a cyclic manner, whereas the increase of CDK4 and 2 levels was sustained. IGF-1 stimulated p21(WAF1/Cip1) expression with a kinetic similar to that of cyclin D1, however p27(Kip1) expression was insensitive to IGF-1. Interestingly, astemizole, a blocker of hEag1 channels, but not E4031, a blocker of HERG channels, inhibited the expression of both cyclins after 6-8h of co-stimulation with IGF-1. However, astemizole failed to modulate CDK4, CDK2, p21(WAF1/Cip1) and p27(Kip1) expression. The down-regulation of hEag1 by siRNA provoked a decrease in cyclin expression. This study is the first to demonstrate that K(+) channels such as hEag1 are directly involved in the IGF-1-induced up-regulation of cyclin D1 and E expression in MCF-7 cells. By identifying more specifically the temporal position of the arrest site induced by the inhibition of hEag1 channels, we confirmed that hEag1 activity is predominantly upstream of the arrest site induced by serum-deprivation, prior to the up-regulation of both cyclins D1 and E. This article is part of a Special Issue entitled: 11th European Symposium on Calcium.


Assuntos
Ciclina D1/metabolismo , Ciclina E/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Fase G1/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Proteínas Oncogênicas/metabolismo , Astemizol/farmacologia , Linhagem Celular Tumoral , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Fase S/efeitos dos fármacos , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
17.
J Cell Physiol ; 227(12): 3837-46, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22495877

RESUMO

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


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Movimento Celular/fisiologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Canais de Cálcio/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Canais de Potássio Éter-A-Go-Go/genética , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Linfonodos/patologia , Manganês , Invasividade Neoplásica , Proteína ORAI1 , Técnicas de Patch-Clamp , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Int J Cancer ; 131(6): E851-61, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22323115

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive forms of cancer with a tendency to invade surrounding healthy tissues, leading to a largely incurable disease. Despite many advances in modern medicine, there is still a lack of early biomarkers as well as efficient therapeutical strategies. The melastatin-related transient receptor potential 7 channel (TRPM7) is a nonselective cation channel that is involved in maintaining Ca(2+) and Mg(2+) homeostasis. It has been recently reported to regulate cell differentiation, proliferation and migration. However, the role of TRPM7 in PDAC progression is far to be understood. In our study, we show that TRPM7 is 13-fold overexpressed in cancer tissues compared to the healthy ones. Furthermore, TRPM7 staining is stronger in tumors with high grade, suggesting a correlation between TRPM7 expression and PDAC progression. Importantly, TRPM7 expression is inversely related to patient survival. In BxPC-3 cell line, dialyzing the cytoplasm during the patch-clamp whole-cell recording with a 0-Mg(2+) solution activated a nonselective current with a strong outward rectification. This cation current is inhibited by intracellular Mg(2+) and by TRPM7 silencing. The downregulation of TRPM7 by small interference RNA dramatically inhibited intracellular Mg(2+) fluorescence and cell migration without affecting cell proliferation, suggesting that TRPM7 contributes to Mg(2+) entry and cell migration. Moreover, external Mg(2+) following TRPM7 silencing fully restored the cell migration. In summary, our results indicate that TRPM7 is involved in the BxPC-3 cell migration via a Mg(2+)-dependent mechanism and may be a potential biomarker of poor prognosis of PDAC.


Assuntos
Adenocarcinoma/patologia , Carcinoma Ductal Pancreático/patologia , Movimento Celular , Neoplasias Pancreáticas/patologia , Canais de Cátion TRPM/fisiologia , Adenocarcinoma/química , Adenocarcinoma/metabolismo , Cálcio/metabolismo , Carcinoma Ductal Pancreático/química , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Humanos , Magnésio/metabolismo , Neoplasias Pancreáticas/química , Neoplasias Pancreáticas/metabolismo , Proteínas Serina-Treonina Quinases , Canais de Cátion TRPM/análise
19.
Cancers (Basel) ; 14(19)2022 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-36230869

RESUMO

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.

20.
Cell Calcium ; 106: 102621, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35905654

RESUMO

Pancreatic stellate cell (PSC) activation is a major event occurring during pancreatic ductal adenocarcinoma (PDAC) development. Up to now mechanisms underlying their activation by mechanical cues such as the elevated tissue pressure in PDAC remain poorly understood. Here we investigate the role of one potential mechano-transducer, TRPC1 ion channel, in PSC activation. Using pre-activated human siTRPC1 and murine TRPC1-KO PSCs, we show that TRPC1 promotes αSMA (α-smooth muscle actin) expression, the main activation marker, in cooperation with the phosphorylated SMAD2, under normal and elevated pressure. Functional studies following TRPC1 silencing demonstrate the dual role of TRPC1 in the modulation of PSC proliferation and IL-6 secretion through the activation of ERK1/2 and SMAD2 pathways. Moreover, pressurization changes the mechanical behavior of PSCs by increasing their cellular stiffness and emitted traction forces in a TRPC1-dependent manner. In summary, these results point to a role of TRPC1 channels in sensing and transducing the characteristic mechanical properties of the PDAC microenvironment in PSCs.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , MAP Quinases Reguladas por Sinal Extracelular , Humanos , Sistema de Sinalização das MAP Quinases , Camundongos , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Células Estreladas do Pâncreas/metabolismo , Proteína Smad2/metabolismo , Canais de Cátion TRPC , Microambiente Tumoral , Neoplasias Pancreáticas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA