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










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 15: 1345488, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38444763

RESUMO

Acute respiratory distress syndrome (ARDS) is characterized by an exacerbated inflammatory response, severe damage to the alveolar-capillary barrier and a secondary infiltration of protein-rich fluid into the airspaces, ultimately leading to respiratory failure. Resolution of ARDS depends on the ability of the alveolar epithelium to reabsorb lung fluid through active transepithelial ion transport, to control the inflammatory response, and to restore a cohesive and functional epithelium through effective repair processes. Interestingly, several lines of evidence have demonstrated the important role of potassium (K+) channels in the regulation of epithelial repair processes. Furthermore, these channels have previously been shown to be involved in sodium/fluid absorption across alveolar epithelial cells, and we have recently demonstrated the contribution of KvLQT1 channels to the resolution of thiourea-induced pulmonary edema in vivo. The aim of our study was to investigate the role of the KCNQ1 pore-forming subunit of KvLQT1 channels in the outcome of ARDS parameters in a model of acute lung injury (ALI). We used a molecular approach with KvLQT1-KO mice challenged with bleomycin, a well-established ALI model that mimics the key features of the exudative phase of ARDS on day 7. Our data showed that KvLQT1 deletion exacerbated the negative outcome of bleomycin on lung function (resistance, elastance and compliance). An alteration in the profile of infiltrating immune cells was also observed in KvLQT1-KO mice while histological analysis showed less interstitial and/or alveolar inflammatory response induced by bleomycin in KvLQT1-KO mice. Finally, a reduced repair rate of KvLQT1-KO alveolar cells after injury was observed. This work highlights the complex contribution of KvLQT1 in the development and resolution of ARDS parameters in a model of ALI.

2.
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.

3.
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
4.
Cell Mol Life Sci ; 79(6): 284, 2022 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-35526196

RESUMO

BACKGROUND AND AIMS: Recent evidences highlight a role of the mitochondria calcium homeostasis in the development of colorectal cancer (CRC). To overcome treatment resistance, we aimed to evaluate the role of the mitochondrial sodium-calcium-lithium exchanger (NCLX) and its targeting in CRC. We also identified curcumin as a new inhibitor of NCLX. METHODS: We examined whether curcumin and pharmacological compounds induced the inhibition of NCLX-mediated mitochondrial calcium (mtCa2+) extrusion, the role of redox metabolism in this process. We evaluated their anti-tumorigenic activity in vitro and in a xenograft mouse model. We analyzed NCLX expression and associations with survival in The Cancer Genome Atlas (TCGA) dataset and in tissue microarrays from 381 patients with microsatellite instability (MSI)-driven CRC. RESULTS: In vitro, curcumin exerted strong anti-tumoral activity through its action on NCLX with mtCa2+ and reactive oxygen species overload associated with a mitochondrial membrane depolarization, leading to reduced ATP production and apoptosis. NCLX inhibition with pharmacological and molecular approaches reproduced the effects of curcumin. NCLX inhibitors decreased CRC tumor growth in vivo. Both transcriptomic analysis of TCGA dataset and immunohistochemical analysis of tissue microarrays demonstrated that higher NCLX expression was associated with MSI status, and for the first time, NCLX expression was significantly associated with recurrence-free survival. CONCLUSIONS: Our findings highlight a novel anti-tumoral mechanism of curcumin through its action on NCLX and mitochondria calcium overload that could benefit for therapeutic schedule of patients with MSI CRC.


Assuntos
Neoplasias Colorretais , Curcumina , Instabilidade de Microssatélites , Trocador de Sódio e Cálcio , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Curcumina/farmacologia , Humanos , Camundongos , Repetições de Microssatélites , Proteínas Mitocondriais/metabolismo , Trocador de Sódio e Cálcio/antagonistas & inibidores
5.
Front Physiol ; 13: 1069466, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36699692

RESUMO

Alveolar ion and fluid absorption is essential for lung homeostasis in healthy conditions as well as for the resorption of lung edema, a key feature of acute respiratory distress syndrome. Liquid absorption is driven by active transepithelial sodium transport, through apical ENaC Na+ channels and basolateral Na+/K+-ATPase. Our previous work unveiled that KvLQT1 K+ channels also participate in the control of Na+/liquid absorption in alveolar epithelial cells. Our aim was to further investigate the function of KvLQT1 channels and their interplay with other channels/transporters involved in ion/liquid transport in vivo using adult wild-type (WT) and KvLQT1 knock-out (KO) mice under physiological conditions and after thiourea-induced lung edema. A slight but significant increase in water lung content (WLC) was observed in naïve KvLQT1-KO mice, relative to WT littermates, whereas lung function was generally preserved and histological structure unaltered. Following thiourea-induced lung edema, KvLQT1-KO did not worsen WLC or lung function. Similarly, lung edema was not aggravated by the administration of a KvLQT1 inhibitor (chromanol). However, KvLQT1 activation (R-L3) significantly reduced WLC in thiourea-challenged WT mice. The benefits of R-L3 were prevented in KO or chromanol-treated WT mice. Furthermore, R-L3 treatment had no effect on thiourea-induced endothelial barrier alteration but restored or enhanced the levels of epithelial alveolar AQP5, Na+/K+-ATPase, and ENaC expressions. Altogether, the results indicate the benefits of KvLQT1 activation in the resolution of lung edema, probably through the observed up-regulation of epithelial alveolar channels/transporters involved in ion/water transport.

6.
Am J Cancer Res ; 11(1): 251-263, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33520372

RESUMO

It's now clearly established that the tumor microenvironment participates to tumor development. Among the different actors contributing to these processes, ion channels, located at the cancer cell surface, play a major role. We recently demonstrated that the association of Kv10.1, Orai1 and SPCA2 is crucial to promote the collagen-induced survival of MCF-7 breast cancer cells. By using siRNA directed against SPCA2, we shown that this protein is involved in the regulation of the activity, the expression and the sub-cellular localization of Kv10.1. In addition, it has been demonstrated that SPCA2 is involved in SICE in MCF-7 cells and that the N- and the C-terminal parts of this protein are necessary to interact and to produce Ca2+ entry. However, no information is available about the necessary SPCA2's important region to regulate Kv10.1. The aim of our work is to evaluate how SPCA2 could interact with Kv10.1 channel to induce survival promotion. By using different SPCA2 mutants, we evaluate the role of the N- and C-terminal sections on the expression and the activity of Kv10.1 channels. In addition, we analyzed the impact of these deletions on the collagen 1-induced cell survival. Our results bring out new information about the regulation of Kv10.1 channel through SPCA2. More specifically how the N- and C-terminus of this Ca2+ transporter regulate Kv10.1 expression, trafficking, and function suggesting new opportunities to target Kv10.1 channels in cancer progression.

7.
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
8.
Biochim Biophys Acta Mol Cell Res ; 1867(4): 118644, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31931022

RESUMO

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.


Assuntos
Canais de Cálcio/metabolismo , Neoplasias/metabolismo , Oxigênio/metabolismo , Canais de Potássio/metabolismo , Microambiente Tumoral , Animais , Hipóxia Celular , Humanos
9.
Mol Pharm ; 16(9): 3814-3822, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31356090

RESUMO

A combination of microbubbles (MBs) and ultrasound (US) is an emerging method for noninvasive and targeted enhancement of anti-cancer drug uptake. This method showed an increase local drug extravasation in tumor tissue while reducing the systemic adverse effects in various tumor models. The present study aims to evaluate the effectiveness of this approach for Nab-paclitaxel delivery in a pancreatic tumor model. US and MBs of different types in combination with Nab-paclitaxel showed a loss in cell viability of pancreatic cancer cells in comparison with Nab-paclitaxel treatment alone in in vitro scenario. The in vivo data revealed that US and MBs in combination with Nab-paclitaxel induced a significant decrease in the tumor volume in a subcutaneous pancreatic adenocarcinoma mouse model in comparison to tumors treated with Nab-paclitaxel alone. The postmortem anatomopathological analyses of tumor tissues partially confirmed these results. In conclusion, this study demonstrates that MB-assisted US is a relevant technology to increase the therapeutic effectiveness of Nab-paclitaxel in a pancreatic cancer model.


Assuntos
Albuminas/uso terapêutico , Antineoplásicos Fitogênicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Meios de Contraste/uso terapêutico , Sistemas de Liberação de Medicamentos/métodos , Microbolhas/uso terapêutico , Nanopartículas/química , Paclitaxel/uso terapêutico , Ultrassonografia/métodos , Animais , Antineoplásicos Fitogênicos/química , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/tratamento farmacológico , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancers (Basel) ; 11(3)2019 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-30884858

RESUMO

In the battle against cancer cells, therapeutic modalities are drastically limited by intrinsic or acquired drug resistance. Resistance to therapy is not only common, but expected: if systemic agents used for cancer treatment are usually active at the beginning of therapy (i.e., 90% of primary breast cancers and 50% of metastases), about 30% of patients with early-stage breast cancer will have recurrent disease. Altered expression of ion channels is now considered as one of the hallmarks of cancer, and several ion channels have been linked to cancer cell resistance. While ion channels have been associated with cell death, apoptosis and even chemoresistance since the late 80s, the molecular mechanisms linking ion channel expression and/or function with chemotherapy have mostly emerged in the last ten years. In this review, we will highlight the relationships between ion channels and resistance to chemotherapy, with a special emphasis on the underlying molecular mechanisms.

11.
Sci Rep ; 9(1): 1175, 2019 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-30718673

RESUMO

In the last years it has been shown that many components of tumor microenvironment (TM) can induce cell signaling that permit to breast cancer cells (BC) to maintain their aggressiveness. Ion channels have a role in mediating TM signal; recently we have demonstrated a functional collaboration between Kv10.1 and Orai1 channels in mediating the pro-survival effect of collagen 1 on BC cells. Here we show how SPCA2 (Secretory Pathway Ca2+ ATPase) has a role in this process and is able to support survival and proliferation induced by collagen 1. By participating to an auto-sustaining loop, SPCA2 enhances membrane expression of Kv10.1 and Orai1; the activity of every component of this trio is necessary to mediate a store independent calcium entry (SICE). This SICE is fundamental to maintain both the activation of the pro-survival pathway and the membrane localization and consequently the activity of the two channels. Moreover, the three proteins and the collagen receptor DDR1 are overexpressed only in aggressive tumors tissues. In this work, we propose a novel association between SPCA2, Kv10.1 and Orai1 involved in mediating transduction signals from TM to the BC cells that can be potentially exploited in the search of novel therapeutic targets specific to tumor tissues.


Assuntos
Neoplasias da Mama/fisiopatologia , ATPases Transportadoras de Cálcio/metabolismo , Sobrevivência Celular , Canais de Potássio Éter-A-Go-Go/metabolismo , Proteína ORAI1/metabolismo , Transporte Biológico , Cálcio/metabolismo , Proliferação de Células , Feminino , Humanos , Células MCF-7 , Modelos Biológicos , Transdução de Sinais , Microambiente Tumoral
12.
Cancer Chemother Pharmacol ; 82(1): 149-157, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29774408

RESUMO

INTRODUCTION: Data suggest a role of the potassium channel SK3 (KCNN3 gene) in oxaliplatin-induced neurotoxicity (OIN). Length variations in the polymorphic CAG repeat of the KCNN3 gene may be associated with the risk of OIN. MATERIALS AND METHODS: We performed patch-clamp experiments on HEK293 cell lines, expressing SK3 channel isoforms with short (11) or long (24) CAG repetitions, to measure intracellular calcium concentrations to test the effects of oxaliplatin on current density. A retrospective study was carried out on patients with colorectal cancer who had received oxaliplatin-based chemotherapy. DNA for KCNN3 genotyping was extracted from leukocytes. The region containing the CAG repeats was amplified by PCR and the products separated by capillary electrophoresis for length analysis. The patients were divided into three groups depending on whether they carried two short alleles, one short allele and one long allele, or two long alleles. The primary endpoint was the onset of grade 2 or 3 neuropathy to oxaliplatin. RESULTS: There was no difference in current density, but oxaliplatin induced a differential effect on apamin-sensitive current density between the two isoforms expressed in the HEK cell lines. There was a significant reduction of store-operated calcium entry into cells expressing the short and more active isoform only after high concentration of oxaliplatin exposition. Eighty-six patients were included in the clinical study. There was no significant association between OIN and KCNN3 polymorphism for the three groups. CONCLUSION: We observed a slight association between OIN and CAG repeat polymorphisms of the KCNN3 gene in a preclinical model, but not a clinical study.


Assuntos
Síndromes Neurotóxicas/genética , Oxaliplatina/efeitos adversos , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Feminino , Fluoruracila/administração & dosagem , Células HEK293 , Humanos , Leucovorina/administração & dosagem , Masculino , Pessoa de Meia-Idade , Síndromes Neurotóxicas/sangue , Síndromes Neurotóxicas/etiologia , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina/administração & dosagem , Técnicas de Patch-Clamp , Polimorfismo Genético , Repetições de Trinucleotídeos
13.
Clin Colorectal Cancer ; 17(1): e109-e113, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29153431

RESUMO

Colorectal cancer is a major problem for public health worldwide because of its frequency and its severity. Many efforts have been carried to target the vascular endothelial growth factor (VEGF) pathway, one of the main promoters of pathological angiogenesis. Therapeutic monoclonal antibodies against VEGF have emerged as essential biopharmaceuticals for the advanced stages of the disease, in association with appropriate backbone chemotherapy. Unfortunately, after an initial benefit for the patients, resistance invariably develops. These mechanisms of resistance are largely studied and recent publications indicate that the interleukin (IL)-17/IL-17 receptor (IL-17R)A axis could be a key player in the pathological progression. In this mini review, we present evidence for IL-17A/IL-17RA axis targeting in colorectal cancer to improve efficiency of anti-VEGF therapy and to implement a new therapeutic strategy.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Interleucina-17/antagonistas & inibidores , Receptores de Interleucina-17/antagonistas & inibidores , Inibidores da Angiogênese/farmacologia , Animais , Anticorpos Monoclonais/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/fisiologia , Humanos , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
14.
BMC Cancer ; 17(1): 220, 2017 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-28347290

RESUMO

BACKGROUND: Colorectal cancer is a major public health issue worldwide. Interleukin-17 (IL-17) and Th17 (T-helper cell type 17)-related molecules are involved in tumor development and in resistance to bevacizumab, an anti-vascular endothelial growth factor monoclonal antibody used in association with chemotherapy in metastatic colorectal cancer. Some studies have previously shown that IL-17A and IL-17F polymorphisms, respectively rs2275913 and rs763780, are associated with gastric or colorectal cancer risk. Here we aimed at studying the influence of IL-17A-related individual factors on overall survival and progression-free survival in patients with metastatic colorectal cancer treated with a bevacizumab-based chemotherapy. METHODS: Pre-treatment serum biomarkers were retrospectively evaluated in 122 metastatic colorectal cancer patients treated by bevacizumab in combination with chemotherapy at 2-weeks intervals in a prospective cohort study (NCT00489697). The polymorphisms of IL-17A and IL-17F were analyzed by polymerase chain reaction - restriction fragment length polymorphism. Serum concentrations of Th17-related cytokines were measured by MultiPlex. The impact of individual parameters on overall survival and progression-free survival was assessed using multivariate Cox models. RESULTS: High baseline IL-17A serum concentrations were significantly associated with shorter progression-free survival [p = 0.043]. Other baseline serum Th17-related cytokines and polymorphisms of IL-17 were not associated with overall survival or progression-free survival. CONCLUSIONS: In this ancillary study, baseline serum IL-17A concentration is the only Th17/IL-17 related factor that was significantly associated with the response of patients with metastatic colorectal cancer to bevacizumab. But this main significant result is highly dependent on one case which, if left out, weakens the data. Other clinical studies are required to confirm this association. TRIAL REGISTRATION: NCT00489697 . June 20, 2007.


Assuntos
Neoplasias Colorretais/sangue , Neoplasias Colorretais/tratamento farmacológico , Predisposição Genética para Doença , Interleucina-17/sangue , Idoso , Bevacizumab/administração & dosagem , Bevacizumab/efeitos adversos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Estudos de Associação Genética , Humanos , Interleucina-17/genética , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Polimorfismo de Nucleotídeo Único
15.
Respir Res ; 16: 100, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26335442

RESUMO

BACKGROUND: Extensive alveolar epithelial injury and remodelling is a common feature of acute lung injury and acute respiratory distress syndrome (ARDS) and it has been established that epithelial regeneration, and secondary lung oedema resorption, is crucial for ARDS resolution. Much evidence indicates that K(+) channels are regulating epithelial repair processes; however, involvement of the KCa3.1 channels in alveolar repair has never been investigated before. RESULTS: Wound-healing assays demonstrated that the repair rates were increased in primary rat alveolar cell monolayers grown on a fibronectin matrix compared to non-coated supports, whereas an anti-ß1-integrin antibody reduced it. KCa3.1 inhibition/silencing impaired the fibronectin-stimulated wound-healing rates, as well as cell migration and proliferation, but had no effect in the absence of coating. We then evaluated a putative relationship between KCa3.1 channel and the migratory machinery protein ß1-integrin, which is activated by fibronectin. Co-immunoprecipitation and immunofluorescence experiments indicated a link between the two proteins and revealed their cellular co-distribution. In addition, we demonstrated that KCa3.1 channel and ß1-integrin membrane expressions were increased on a fibronectin matrix. We also showed increased intracellular calcium concentrations as well as enhanced expression of TRPC4, a voltage-independent calcium channel belonging to the large TRP channel family, on a fibronectin matrix. Finally, wound-healing assays showed additive effects of KCa3.1 and TRPC4 inhibitors on alveolar epithelial repair. CONCLUSION: Taken together, our data demonstrate for the first time complementary roles of KCa3.1 and TRPC4 channels with extracellular matrix and ß1-integrin in the regulation of alveolar repair processes.


Assuntos
Células Epiteliais Alveolares/metabolismo , Integrina beta1/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Alvéolos Pulmonares/metabolismo , Cicatrização , Células Epiteliais Alveolares/efeitos dos fármacos , Células Epiteliais Alveolares/patologia , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Relação Dose-Resposta a Droga , Fibronectinas/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Masculino , Bloqueadores dos Canais de Potássio/farmacologia , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/patologia , Interferência de RNA , Ratos Sprague-Dawley , Transdução de Sinais , Canais de Cátion TRPC/antagonistas & inibidores , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Fatores de Tempo , Transfecção , Cicatrização/efeitos dos fármacos
16.
Am J Physiol Cell Physiol ; 306(4): C307-19, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24196531

RESUMO

Efficient repair of epithelial tissue, which is frequently exposed to insults, is necessary to maintain its functional integrity. It is therefore necessary to better understand the biological and molecular determinants of tissue regeneration and to develop new strategies to promote epithelial repair. Interestingly, a growing body of evidence indicates that many members of the large and widely expressed family of K(+) channels are involved in regulation of cell migration and proliferation, key processes of epithelial repair. First, we briefly summarize the complex mechanisms, including cell migration, proliferation, and differentiation, engaged after epithelial injury. We then present evidence implicating K(+) channels in the regulation of these key repair processes. We also describe the mechanisms whereby K(+) channels may control epithelial repair processes. In particular, changes in membrane potential, K(+) concentration, cell volume, intracellular Ca(2+), and signaling pathways following modulation of K(+) channel activity, as well as physical interaction of K(+) channels with the cytoskeleton or integrins are presented. Finally, we discuss the challenges to efficient, specific, and safe targeting of K(+) channels for therapeutic applications to improve epithelial repair in vivo.


Assuntos
Movimento Celular , Proliferação de Células , Células Epiteliais/metabolismo , Canais de Potássio/metabolismo , Potássio/metabolismo , Regeneração , Cicatrização , Animais , Humanos , Potenciais da Membrana , Transdução de Sinais
17.
Int J Oncol ; 44(3): 838-48, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24366043

RESUMO

K+ channels, which are overexpressed in several cancers, have been identified as regulators of cell proliferation and migration, key processes in cancer development/propagation. Their role in lung cancer has not been studied extensively; but we showed previously that KvLQT1 channels are involved in cell migration, proliferation and repair of normal lung epithelial cells. We now investigated the role of these channels in lung cancer cell lines and their expression levels in human lung adenocarcinoma (AD) tissues. First, we observed that the wound-healing rates of A549 lung adenocarcinoma cell monolayers were reduced by clofilium and chromanol or after silencing with KvLQT1-specific siRNA. Dose-dependent decrease of A549 cell growth and cell accumulation in G0/G1 phase were seen after KvLQT1 inhibition. Clofilium also affected 2D and 3D migration of A549 cells. Similarly, H460 cell growth, migration and wound healing were diminished by this drug. Because K+ channel overexpression has been encountered in some cancers, we assessed KvLQT1 expression levels in tumor tissues from patients with lung AD. KvLQT1 protein expression in tumor samples was increased by 1.5- to 7-fold, compared to paired non-neoplastic tissues, in 17 of 26 patients. In summary, our data reveal that KvLQT1 channel blockade efficiently reduces A549 and H460 cell proliferation and migration. Moreover, KvLQT1 overexpression in AD samples suggests that it could be a potential therapeutic target in lung cancer.


Assuntos
Adenocarcinoma/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Canais de Potássio KCNQ/genética , Neoplasias Pulmonares/genética , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Carcinogênese , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Canais de Potássio KCNQ/biossíntese , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Compostos de Amônio Quaternário/administração & dosagem , RNA Interferente Pequeno/genética , Cicatrização/genética
18.
Cancer Res ; 73(15): 4852-61, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23774210

RESUMO

The SK3 channel, a potassium channel, was recently shown to control cancer cell migration, a critical step in metastasis outgrowth. Here, we report that expression of the SK3 channel was markedly associated with bone metastasis. The SK3 channel was shown to control constitutive Ca(2+) entry and cancer cell migration through an interaction with the Ca(2+) channel Orai1. We found that the SK3 channel triggers an association with the Orai1 channel within lipid rafts. This localization of an SK3-Orai1 complex seemed essential to control cancer cell migration. This suggests that the formation of this complex in lipid rafts is a gain-of-function, because we showed that none of the individual proteins were able to promote the complete phenotype. We identified the alkyl-lipid Ohmline as a disrupting agent for SK3-Orai1 lipid raft localization. Upon Ohmline treatment, the SK3-Orai1 complex moved away from lipid rafts, and SK3-dependent Ca(2+) entry, migration, and bone metastases were subsequently impaired. The colocalization of SK3 and Orai1 in primary human tumors and bone metastases further emphasized the clinical relevance of our observations. Targeting SK3-Orai1 in lipid rafts may inaugurate innovative approaches to inhibit bone metastases.


Assuntos
Neoplasias Ósseas/metabolismo , Canais de Cálcio/metabolismo , Microdomínios da Membrana/metabolismo , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/secundário , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Animais , Western Blotting , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Movimento Celular , Modelos Animais de Doenças , Feminino , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Invasividade Neoplásica/patologia , Proteína ORAI1 , Técnicas de Patch-Clamp
19.
Biochem Biophys Res Commun ; 397(1): 42-7, 2010 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-20519134

RESUMO

Lost of adenomatous polyposis coli gene (Apc) disturbs the migration of intestinal epithelial cells but the mechanisms have not been fully characterized. Since we have demonstrated that SK3/KCa2.3 channel promotes cancer cell migration, we hypothesized that Apc mutation may affect SK3/KCa2.3 channel-mediated colon epithelial cell motility. We report evidence that SK3/KCa2.3 channel promotes colon epithelial cells motility. Following Apc mutation SK3/KCa2.3 expression is largely reduced leading to a suppression of the SK3/KCa2.3 channel mediated-cell migration. Our findings reveal a previously unknown function of the SK3/KCa2.3 channel in epithelial colonic cells, and suggest that Apc is a powerful regulator SK3/KCa2.3 channel.


Assuntos
Proteína da Polipose Adenomatosa do Colo/metabolismo , Movimento Celular , Colo/metabolismo , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Linhagem Celular , Colo/citologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Mutantes , Canais de Potássio Ativados por Cálcio de Condutância Baixa
20.
Exp Cell Res ; 315(20): 3620-30, 2009 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-19646982

RESUMO

Cell migration and invasion are required for tumour cells to spread from the primary tumour bed so as to form secondary tumours at distant sites. We report evidence of an unusual expression of KCa2.3 (SK3) protein in melanoma cell lines but not in normal melanocytes. Knockdown of the KCa2.3 channel led to plasma membrane depolarization, decreased 2D and 3D cell motility. Conversely, enforced production of KCa2.3 protein in KCa2.3 non-expressing cells led to the plasma membrane becoming hyperpolarized, and enhanced cell motility. In contrast, KCa3.1 channels had no effect on cell motility despite an active role in regulating membrane potential. Our data also suggest that membrane hyperpolarization increases melanoma cell motility and that this occurs through the KCa2.3 channel. Our findings reveal a previously unknown function of the KCa2.3 channel, and suggest that the KCa2.3 channel might be the only member of the Ca(2+)-activated K(+) channel family involved in melanoma cell motility pathways.


Assuntos
Movimento Celular/fisiologia , Melanoma/metabolismo , Melanoma/patologia , Potenciais da Membrana/fisiologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/fisiologia , Apamina/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Clotrimazol/farmacologia , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Expressão Gênica/genética , Humanos , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/fisiologia , Melanócitos/citologia , Melanócitos/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Pirazóis/farmacologia , RNA Antissenso/genética , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...