Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Strahlenther Onkol ; 193(6): 466-471, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28396899

RESUMO

PURPOSE: The antiapoptotic B­cell lymphoma 2 (BCL2) gene is a key player in cancer development and progression. A functional single-nucleotide polymorphism (c.-938C>A, rs2279115) in the inhibitory P2 BCL2 gene promoter has been associated with clinical outcomes in various types of cancer. Aim of the present study was to analyze the role of BCL2-938C>A genotypes in prostate cancer mortality. METHODS: The association between BCL2-938C>A (rs2279115) genotypes and prostate cancer outcome was studied within the prospective PROCAGENE study comprising 702 prostate cancer patients. RESULTS: During a median follow-up time of 92 months, 120 (17.1%) patients died. A univariate Cox regression model showed a significant association of the CC genotype with reduced cancer-specific survival (CSS; hazard ratio, HR, 2.13, 95% confidence interval, CI, 1.10-4.12; p = 0.024) and overall survival (OS; HR 2.34, 95% CI 1.58-3.47; p < 0.001). In a multivariate Cox regression model including age at diagnosis, risk group, and androgen deprivation therapy, the CC genotype remained a significant predictor of poor CSS (HR 2.05, 95% CI 1.05-3.99; p = 0.034) and OS (HR 2.25, 95% CI 1.51-3.36; p < 0.001). CONCLUSION: This study provides evidence that the homozygous BCL2-938 CC genotype is associated with OS and C in prostate cancer patients.


Assuntos
Genótipo , Neoplasias da Próstata/genética , Neoplasias da Próstata/mortalidade , Proteínas Proto-Oncogênicas c-bcl-2/genética , Idoso , Alelos , Antagonistas de Androgênios/uso terapêutico , Terapia Combinada , Seguimentos , Homozigoto , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Gradação de Tumores , Estadiamento de Neoplasias , Polimorfismo de Nucleotídeo Único/genética , Modelos de Riscos Proporcionais , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Radioterapia Conformacional , Análise de Sobrevida
2.
Adv Exp Med Biol ; 993: 505-521, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28900930

RESUMO

TRPC channels have been suggested as potential candidates mediating store-operated Ca2+ entry (SOCE) in cardiomyocytes. There is increasing evidence that the TRPC isoforms TRPC1 and TRPC4 might fulfill the function as SOCs, in concert with or in parallel to the key players of SOCE, Orai1, and STIM1. Several other isoforms, e.g., TRPC3, TRPC6, and TRPC7, might rather associate to receptor-activated diacylglycerol (DAG)-sensitive ion channels. However, the exact activation mode has not been elucidated yet, given the characteristic of TRPC channels to heteromerize to unpredictable ion channel assemblies. Despite the incomplete information about TRPC activation, there is common agreement that they are crucial Ca2+ components in cardiac signaling and disease. All TRPC isoforms, TRPC1, TRPC3, TRPC4, TRPC5, TRPC6, and TRPC7, are differentially regulated in cardiac disease, and nearly all of them have been shown to impact cardiac signaling pathways that accelerate cardiac disease development. In particular, the calcineurin-nuclear factor of activated T-cell (NFAT) signaling pathway has repeatedly been linked to a TRPC-dependent Ca2+ influx in cardiomyocytes. Moreover, the protein kinases PKG and PKC have been found to modulate TRPC function and the hypertrophic response. Other signaling molecules, such as the serine/threonine kinase Ca2+/calmodulin-dependent protein kinase II (CamKII) or the oxidative stress molecule, NADPH oxidase 2 (NOX2), have also been related to TRPC-dependent effects in the heart.The present chapter provides a comprehensive overview of TRPC channels as Ca2+ entities in cardiomyocytes, their interplay with Ca2+ signaling pathways, and role in cardiac pathology.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Cardiopatias/metabolismo , Coração/fisiologia , Canais de Cátion TRPC/metabolismo , Animais , Humanos
3.
Circ Res ; 114(2): 249-256, 2014 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-24218169

RESUMO

RATIONALE: The Na+ / K+ ATPase (NKA) directly regulates intracellular Na+ levels, which in turn indirectly regulates Ca2+ levels by proximally controlling flux through the Na+ / Ca2+ exchanger (NCX1). Elevated Na+ levels have been reported during heart failure, which permits some degree of reverse-mode Ca2+ entry through NCX1, as well as less efficient Ca2+ clearance. OBJECTIVE: To determine whether maintaining lower intracellular Na+ levels by NKA overexpression in the heart would enhance forward-mode Ca2+ clearance and prevent reverse-mode Ca2+ entry through NCX1 to protect the heart. METHODS AND RESULTS: Cardiac-specific transgenic mice overexpressing either NKA-α1 or NKA-α2 were generated and subjected to pressure overload hypertrophic stimulation. We found that although increased expression of NKA-α1 had no protective effect, overexpression of NKA-α2 significantly decreased cardiac hypertrophy after pressure overload in mice at 2, 10, and 16 weeks of stimulation. Remarkably, total NKA protein expression and activity were not altered in either of these 2 transgenic models because increased expression of one isoform led to a concomitant decrease in the other endogenous isoform. NKA-α2 overexpression but not NKA-α1 led to significantly faster removal of bulk Ca2+ from the cytosol in a manner requiring NCX1 activity. Mechanistically, overexpressed NKA-α2 showed greater affinity for Na+ compared with NKA-α1, leading to more efficient clearance of this ion. Furthermore, overexpression of NKA-α2 but not NKA-α1 was coupled to a decrease in phospholemman expression and phosphorylation, which would favor greater NKA activity, NCX1 activity, and Ca2+ removal. CONCLUSIONS: Our results suggest that the protective effect produced by increased expression of NKA-α2 on the heart after pressure overload is due to more efficient Ca2+ clearance because this isoform of NKA preferentially enhances NCX1 activity compared with NKA-α1.


Assuntos
Cardiomegalia/prevenção & controle , Miocárdio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Remodelação Ventricular , Animais , Cálcio/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Cardiomegalia/enzimologia , Cardiomegalia/genética , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Células Cultivadas , Modelos Animais de Doenças , Isoenzimas , Camundongos , Camundongos Transgênicos , Miocárdio/patologia , Fosforilação , Ratos , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , Fatores de Tempo , Transfecção , Regulação para Cima
4.
Pflugers Arch ; 466(6): 1151-62, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24510065

RESUMO

Among the myriad of molecular alterations occurring in heart failure development, aggravation of the disease is often attributed to global or local changes in protein kinase activity, thus making protein kinases attractive targets for therapeutic intervention. Since protein kinases do not only have maladaptive roles, but also contribute to the physiological integrity of cells, it is a challenging task to circumvent undesired inhibition of protein kinase activity. Identification of posttranslational modifications and/or protein-protein interactions that are exclusively apparent under pathophysiological conditions provides exciting information for alternative non-kinase inhibitory treatment strategies that eliminate maladaptive functions of a protein kinase, but preserve the beneficial ones. Here, we focus on the disease-specific regulation of a number of protein kinases, namely, Ca(2+)/calmodulin-dependent protein kinase II isoform δ (CaMKIIδ), G protein-coupled receptor kinase 2 (GRK2), extracellular signal-regulated kinase 1 and 2 (ERK1/2), protein kinase D (PKD) and protein kinase C isoform ß2 (PKCß2), which are embedded in complex signal transduction pathways implicated in heart failure development, and discuss potential avenues for novel treatment strategies to combat heart disease.


Assuntos
Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Quinase 2 de Receptor Acoplado a Proteína G/metabolismo , Insuficiência Cardíaca/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína Quinase C/metabolismo , Animais , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Insuficiência Cardíaca/enzimologia , Humanos
5.
Circ Res ; 108(2): 265-72, 2011 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-21252153

RESUMO

Transient receptor potential (TRP) channels of multiple subclasses are expressed in the heart, although their functions are only now beginning to emerge, especially for the TRPC subclass that appears to regulate the cardiac hypertrophic response. Although TRP channels permeate many different cations, they are most often ascribed a specific biological function because of Ca(2+) influx, either for microdomain signaling or to reload internal Ca(2+) stores in the endoplasmic reticulum through a store-operated mechanism. However, adult cardiac myocytes arguably do not require store-operated Ca(2+) entry to regulate sarcoplasmic reticulum Ca(2+) levels and excitation-contraction coupling; hence, TRP channels expressed in the heart most likely coordinate signaling within local domains or through direct interaction with Ca(2+)-dependent regulatory proteins. Here, we review the emerging evidence that TRP channels, especially TRPCs, are critical regulators of microdomain signaling in the heart to control pathological hypertrophy in coordination with signaling through effectors such as calcineurin and NFAT (nuclear factor of activated T cells).


Assuntos
Cardiomegalia/fisiopatologia , Canais de Cátion TRPC/fisiologia , Animais , Calcineurina/fisiologia , Cálcio/metabolismo , Humanos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/fisiologia , Transdução de Sinais/fisiologia
6.
Proc Natl Acad Sci U S A ; 107(15): 7000-5, 2010 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-20351294

RESUMO

Pathologic hypertrophy of the heart is regulated through membrane-bound receptors and intracellular signaling pathways that function, in part, by altering Ca(2+) handling and Ca(2+)-dependent signaling effectors. Transient receptor potential canonical (TRPC) channels are important mediators of Ca(2+)-dependent signal transduction that can sense stretch or activation of membrane-bound receptors. Here we generated cardiac-specific transgenic mice that express dominant-negative (dn) TRPC3, dnTRPC6, or dnTRPC4 toward blocking the activity of the TRPC3/6/7 or TRPC1/4/5 subfamily of channels in the heart. Remarkably, all three dn transgenic strategies attenuated the cardiac hypertrophic response following either neuroendocrine agonist infusion or pressure-overload stimulation. dnTRPC transgenic mice also were partially protected from loss of cardiac functional performance following long-term pressure-overload stimulation. Importantly, adult myocytes isolated from hypertrophic WT hearts showed a unique Ca(2+) influx activity under store-depleted conditions that was not observed in myocytes from hypertrophied dnTRPC3, dnTRPC6, or dnTRPC4 hearts. Moreover, dnTRPC4 inhibited the activity of the TRPC3/6/7 subfamily in the heart, suggesting that these two subfamilies function in coordinated complexes. Mechanistically, inhibition of TRPC channels in transgenic mice or in cultured neonatal myocytes significantly reduced activity in the calcineurin-nuclear factor of activated T cells (NFAT), a known Ca(2+)-dependent hypertrophy-inducing pathway. Thus, TRPC channels are necessary mediators of pathologic cardiac hypertrophy, in part through a calcineurin-NFAT signaling pathway.


Assuntos
Cardiomegalia/patologia , Canais de Potencial de Receptor Transitório/fisiologia , Animais , Calcineurina/metabolismo , Cardiomegalia/metabolismo , Ecocardiografia/métodos , Genes Dominantes , Camundongos , Camundongos Transgênicos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Regiões Promotoras Genéticas , Transdução de Sinais , Canais de Cátion TRPC/metabolismo , Canal de Cátion TRPC6 , Canais de Potencial de Receptor Transitório/metabolismo
7.
FEBS Lett ; 582(18): 2696-702, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18602918

RESUMO

VEGF-induced Ca2+ signalling was investigated in CD133+/VEGFR-2+ progenitor cells isolated from human adipose stroma. Colonies derived from CD133+ immunoselected cells displayed inhomogenous Ca2+ signals, with variable magnitude of VEGF-induced Ca2+ entry, which positively correlated with expression of the Ca2+ channel protein TRPC3. High levels of VEGF-induced Ca2+ entry and TRPC3 expression were preferentially detected in rim areas of expanding colonies. Dominant negative suppression of TRPC3 inhibited VEGF-induced Ca2+ entry into CD133+ cells. Our results identify TRPC3 as a key Ca2+ entry channel in a subset of CD133+ stem cells. We suggest TRPC3 as an essential determinant of cell fate in CD133+ progenitor-derived colonies.


Assuntos
Tecido Adiposo/citologia , Antígenos CD/biossíntese , Canais de Cálcio/biossíntese , Glicoproteínas/biossíntese , Células-Tronco/citologia , Canais de Cátion TRPC/biossíntese , Fator A de Crescimento do Endotélio Vascular/metabolismo , Antígeno AC133 , Tecido Adiposo/efeitos dos fármacos , Tecido Adiposo/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Canais de Cálcio/genética , Diferenciação Celular , Linhagem Celular , Células Cultivadas , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neovascularização Fisiológica , Peptídeos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Canais de Cátion TRPC/genética , Fator A de Crescimento do Endotélio Vascular/farmacologia
8.
PLoS One ; 11(12): e0168446, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27992507

RESUMO

The Transient Receptor Potential Channel Subunit 4 (TRPC4) has been considered as a crucial Ca2+ component in cardiomyocytes promoting structural and functional remodeling in the course of pathological cardiac hypertrophy. TRPC4 assembles as homo or hetero-tetramer in the plasma membrane, allowing a non-selective Na+ and Ca2+ influx. Gαq protein-coupled receptor (GPCR) stimulation is known to increase TRPC4 channel activity and a TRPC4-mediated Ca2+ influx which has been regarded as ideal Ca2+ source for calcineurin and subsequent nuclear factor of activated T-cells (NFAT) activation. Functional properties of TRPC4 are also based on the expression of the TRPC4 splice variants TRPC4α and TRPC4ß. Aim of the present study was to analyze cytosolic Ca2+ signals, signaling, hypertrophy and vitality of cardiomyocytes in dependence on the expression level of either TRPC4α or TRPC4ß. The analysis of Ca2+ transients in neonatal rat cardiomyocytes (NRCs) showed that TRPC4α and TRPC4ß affected Ca2+ cycling in beating cardiomyocytes with both splice variants inducing an elevation of the Ca2+ transient amplitude at baseline and TRPC4ß increasing the Ca2+ peak during angiotensin II (Ang II) stimulation. NRCs infected with TRPC4ß (Ad-C4ß) also responded with a sustained Ca2+ influx when treated with Ang II under non-pacing conditions. Consistent with the Ca2+ data, NRCs infected with TRPC4α (Ad-C4α) showed an elevated calcineurin/NFAT activity and a baseline hypertrophic phenotype but did not further develop hypertrophy during chronic Ang II/phenylephrine stimulation. Down-regulation of endogenous TRPC4α reversed these effects, resulting in less hypertrophy of NRCs at baseline but a markedly increased hypertrophic enlargement after chronic agonist stimulation. Ad-C4ß NRCs did not exhibit baseline calcineurin/NFAT activity or hypertrophy but responded with an increased calcineurin/NFAT activity after GPCR stimulation. However, this effect was not translated into an increased propensity towards hypertrophy but rather less hypertrophy during GPCR stimulation. Further analyses revealed that, although hypertrophy was preserved in Ad-C4α NRCs and even attenuated in Ad-C4ß NRCs, cardiomyocytes had an increased apoptosis rate and thus were less viable after chronic GPCR stimulation. These findings suggest that TRPC4α and TRPC4ß differentially affect Ca2+ signals, calcineurin/NFAT signaling and hypertrophy but similarly impair cardiomyocyte viability during GPCR stimulation.


Assuntos
Cardiomegalia/metabolismo , Miócitos Cardíacos/citologia , Canais de Cátion TRPC/genética , Canais de Cátion TRPC/metabolismo , Processamento Alternativo , Animais , Animais Recém-Nascidos , Calcineurina/metabolismo , Cálcio/metabolismo , Cardiomegalia/genética , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Camundongos , Miócitos Cardíacos/metabolismo , Fatores de Transcrição NFATC/metabolismo , Ratos , Transdução de Sinais
9.
Channels (Austin) ; 2(2): 94-9, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18849655

RESUMO

Nonselective and lipid-regulated cation channels formed by TRPC3, TRPC6 and TRPC7 have recently obtained attention in view their potential pathophysiological impact. It appears as a particular challenge to understand the molecular basis of TRPC3/6/7-related diseases in order to further delineate their value as therapeutic targets. The multifunctional nature of these channel proteins, based on a complex, versatile heteromerization potential along with highly promiscuous gating and mixed cation permeation properties, make these channels pivotal players in cellular signaling networks. Here we summarize newsworthy aspects of TRPC3/6/7 physiology and pathophysiology that open the view on novel therapeutic strategies based on these TRPCs as target structures.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Canais de Cátion TRPC/fisiologia , Descoberta de Drogas , Humanos , Canais de Cátion TRPC/efeitos dos fármacos , Canal de Cátion TRPC6
10.
J Biol Chem ; 283(12): 8014-22, 2008 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-18187424

RESUMO

STIM1 and ORAI1 (also termed CRACM1) are essential components of the classical calcium release-activated calcium current; however, the mechanism of the transmission of information of STIM1 to the calcium release-activated calcium/ORAI1 channel is as yet unknown. Here we demonstrate by Förster resonance energy transfer microscopy a dynamic coupling of STIM1 and ORAI1 that culminates in the activation of Ca(2+) entry. Förster resonance energy transfer imaging of living cells provided insight into the time dependence of crucial events of this signaling pathway comprising Ca(2+) store depletion, STIM1 multimerization, and STIM1-ORAI1 interaction. Accelerated store depletion allowed resolving a significant time lag between STIM1-STIM1 and STIM1-ORAI1 interactions. Store refilling reversed both STIM1 multimerization and STIM1-ORAI1 interaction. The cytosolic STIM1 C terminus itself was able, in vitro as well as in vivo, to associate with ORAI1 and to stimulate channel function, yet without ORAI1-STIM1 cluster formation. The dynamic interaction occurred via the C terminus of ORAI1 that includes a putative coiled-coil domain structure. An ORAI1 C terminus deletion mutant as well as a mutant (L273S) with impeded coiled-coil domain formation lacked both interaction as well as functional communication with STIM1 and failed to generate Ca(2+) inward currents. An N-terminal deletion mutant of ORAI1 as well as the ORAI1 R91W mutant linked to severe combined immune deficiency syndrome was similarly impaired in terms of current activation despite being able to interact with STIM1. Hence, the C-terminal coiled-coil motif of ORAI1 represents a key domain for dynamic coupling to STIM1.


Assuntos
Canais de Cálcio/metabolismo , Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Motivos de Aminoácidos/fisiologia , Substituição de Aminoácidos , Canais de Cálcio/genética , Linhagem Celular , Citoplasma/genética , Citoplasma/metabolismo , Humanos , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Proteína ORAI1 , Ligação Proteica/fisiologia , Estrutura Terciária de Proteína/fisiologia , Molécula 1 de Interação Estromal
11.
J Biol Chem ; 281(19): 13588-13595, 2006 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-16537542

RESUMO

Canonical transient receptor potential proteins (TRPC) have been proposed to form homo- or heteromeric cation channels in a variety of tissues, including the vascular endothelium. Assembly of TRPC multimers is incompletely understood. In particular, heteromeric assembly of distantly related TRPC isoforms is still a controversial issue. Because we have previously suggested TRPC proteins as the basis of the redox-activated cation conductance of porcine aortic endothelial cells (PAECs), we set out to analyze the TRPC subunit composition of endogenous endothelial TRPC channels and report here on a redox-sensitive TRPC3-TRPC4 channel complex. The ability of TRPC3 and TRPC4 proteins to associate and to form a cation-conducting pore complex was supported by four lines of evidence as follows: 1) Co-immunoprecipitation experiments in PAECs and in HEK293 cells demonstrated the association of TRPC3 and TRPC4 in the same complex. 2) Fluorescence resonance energy transfer analysis demonstrated TRPC3-TRPC4 association, involving close proximity between the N terminus of TRPC4 and the C terminus of TRPC3 subunits. 3) Co-expression of TRPC3 and TRPC4 in HEK293 cells generated a channel that displayed distinct biophysical and regulatory properties. 4) Expression of dominant-negative TRPC4 proteins suppressed TRPC3-related channel activity in the HEK293 expression system and in native endothelial cells. Specifically, an extracellularly hemagglutinin (HA)-tagged TRPC4 mutant, which is sensitive to blockage by anti-HA-antibody, was found to transfer anti-HA sensitivity to both TRPC3-related currents in the HEK293 expression system and the redox-sensitive cation conductance of PAECs. We propose TRPC3 and TRPC4 as subunits of native endothelial cation channels that are governed by the cellular redox state.


Assuntos
Células Endoteliais/metabolismo , Canais de Cátion TRPC/química , Canais de Cátion TRPC/metabolismo , Animais , Linhagem Celular , Regulação da Expressão Gênica , Humanos , Potenciais da Membrana , Camundongos , Oxirredução , Estresse Oxidativo , Ligação Proteica , Estrutura Quaternária de Proteína , Suínos , Canais de Cátion TRPC/genética
12.
Pflugers Arch ; 451(1): 99-104, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15924237

RESUMO

Ion channels formed by canonical transient receptor potential (TRPC) proteins are considered to be key players in cellular Ca(2+) homeostasis. As permeation of Ca(2+) through TRPC homo- and/or heteromeric channels has been repeatedly demonstrated, analysis of the physiological role of TRPC proteins was so far based on the concept that these proteins form regulated Ca(2+) entry channels. The well-recognized lack of cation selectivity of TRPC channels and the ability to generate substantial monovalent conductances that govern membrane potential and cation gradients were barely appreciated as a physiologically relevant issue. Nonetheless, recent studies suggest monovalent, specifically Na(+) permeation through TRPC cation channels as an important event in TRPC signaling. TRPC-mediated Na(+) entry may be converted into a distinct pattern of cellular Ca(2+) signals by interaction with Na(+)/Ca(2+) exchanger proteins. This review discusses current concepts regarding the link between Na(+) entry through TRPC channels and cellular Ca(2+) signaling.


Assuntos
Cálcio/metabolismo , Trocador de Sódio e Cálcio/fisiologia , Sódio/metabolismo , Canais de Cátion TRPC/fisiologia , Animais , Sinalização do Cálcio , Humanos , Modelos Biológicos , Ratos , Transdução de Sinais/fisiologia
13.
J Biol Chem ; 279(14): 13696-704, 2004 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-14736881

RESUMO

TRPC3 has been suggested as a key component of phospholipase C-dependent Ca(2+) signaling. Here we investigated the role of TRPC3-mediated Na(+) entry as a determinant of plasmalemmal Na(+)/Ca(2+) exchange. Ca(2+) signals generated by TRPC3 overexpression in HEK293 cells were found to be dependent on extracellular Na(+), in that carbachol-stimulated Ca(2+) entry into TRPC3 expressing cells was significantly suppressed when extracellular Na(+) was reduced to 5 mm. Moreover, KB-R9743 (5 microm) an inhibitor of the Na(+)/Ca(2+) exchanger (NCX) strongly suppressed TRPC3-mediated Ca(2+) entry but not TRPC3-mediated Na(+) currents. NCX1 immunoreactivity was detectable in HEK293 as well as in TRPC3-overexpressing HEK293 cells, and reduction of extracellular Na(+) after Na(+) loading with monensin resulted in significant rises in intracellular free Ca(2+) (Ca(2+)(i)) of HEK293 cells. Similar rises in Ca(2+)(i) were recorded in TRPC3-overexpressing cells upon the reduction of extracellular Na(+) subsequent to stimulation with carbachol. These increases in Ca(2+)(i) were associated with outward membrane currents at positive potentials and inhibited by KB-R7943 (5 microm), chelation of extracellular Ca(2+), or dominant negative suppression of TRPC3 channel function. This suggests that Ca(2+) entry into TRPC3-expressing cells involves reversed mode Na(+)/Ca(2+) exchange. Cell fractionation experiments demonstrated co-localization of TRPC3 and NCX1 in low density membrane fractions, and co-immunoprecipitation experiments provided evidence for association of TRPC3 and NCX1. Glutathione S-transferase pull-down experiments revealed that NCX1 interacts with the cytosolic C terminus of TRPC3. We suggest functional and physical interaction of nonselective TRPC cation channels with NCX proteins as a novel principle of TRPC-mediated Ca(2+) signaling.


Assuntos
Sinalização do Cálcio/fisiologia , Canais Iônicos/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Sódio/metabolismo , Tioureia/análogos & derivados , Antiarrítmicos/farmacologia , Cálcio/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Homeostase , Humanos , Rim/citologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Trocador de Sódio e Cálcio/antagonistas & inibidores , Canais de Cátion TRPC , Tioureia/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA