Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Biol Chem ; 289(10): 6372-6382, 2014 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-24464579

RESUMEN

Transient receptor potential canonical (TRPC) channels mediate a critical part of the receptor-evoked Ca(2+) influx. TRPCs are gated open by the endoplasmic reticulum Ca(2+) sensor STIM1. Here we asked which stromal interaction molecule 1 (STIM1) and TRPC domains mediate the interaction between them and how this interaction is used to open the channels. We report that the STIM1 Orai1-activating region domain of STIM1 interacts with the TRPC channel coiled coil domains (CCDs) and that this interaction is essential for opening the channels by STIM1. Thus, disruption of the N-terminal (NT) CCDs by triple mutations eliminated TRPC surface localization and reduced binding of STIM1 to TRPC1 and TRPC5 while increasing binding to TRPC3 and TRPC6. Single mutations in TRPC1 NT or C-terminal (CT) CCDs reduced interaction and activation of TRPC1 by STIM1. Remarkably, single mutations in the TRPC3 NT CCD enhanced interaction and regulation by STIM1. Disruption in the TRPC3 CT CCD eliminated regulation by STIM1 and the enhanced interaction caused by NT CCD mutations. The NT CCD mutations converted TRPC3 from a TRPC1-dependent to a TRPC1-independent, STIM1-regulated channel. TRPC1 reduced the FRET between BFP-TRPC3 and TRPC3-YFP and between CFP-TRPC3-YFP upon stimulation. Accordingly, knockdown of TRPC1 made TRPC3 STIM1-independent. STIM1 dependence of TRPC3 was reconstituted by the TRPC1 CT CCD alone. Knockout of Trpc1 and Trpc3 similarly inhibited Ca(2+) influx, and inhibition of Trpc3 had no further effect on Ca(2+) influx in Trpc1(-/-) cells. Cell stimulation enhanced the formation of Trpc1-Stim1-Trpc3 complexes. These findings support a model in which the TRPC3 NT and CT CCDs interact to shield the CT CCD from interaction with STIM1. The TRPC1 CT CCD dissociates this interaction to allow the STIM1 Orai1-activating region within STIM1 access to the TRPC3 CT CCD and regulation of TRPC3 by STIM1. These studies provide evidence that the TRPC channel CCDs participate in channel gating.


Asunto(s)
Activación del Canal Iónico , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Dominios y Motivos de Interacción de Proteínas , Canales Catiónicos TRPC/metabolismo , Animales , Canales de Calcio/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Proteína ORAI1 , Interferencia de ARN , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/química , Canales Catiónicos TRPC/genética
2.
J Biol Chem ; 286(36): 31799-809, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21768109

RESUMEN

Both TRPC6 and reactive oxygen species (ROS) play an important role in regulating vascular function. However, their interplay has not been explored. The present study examined whether activation of TRPC6 in vascular smooth muscle cells (VSMCs) by ROS was a physiological mechanism for regulating vascular tone by vasoconstrictors. In A7r5 cells, arginine vasopressin (AVP) evoked a striking Ca(2+) entry response that was significantly attenuated by either knocking down TRPC6 using siRNA or inhibition of NADPH oxidases with apocynin or diphenyleneiodonium. Inhibition of TRPC6 or ROS production also decreased AVP-stimulated membrane currents. In primary cultured aortic VSMCs, catalase and diphenyleneiodonium significantly suppressed AVP- and angiotensin II-induced whole cell currents and Ca(2+) entry, respectively. In freshly isolated and endothelium-denuded thoracic aortas, hyperforin (an activator of TRPC6), but not its vehicle, induced dose- and time-dependent constriction in TRPC6 wide type (WT) mice. This response was not observed in TRPC6 knock-out (KO) mice. Consistent with the ex vivo study, hyperforin stimulated a robust Ca(2+) entry in the aortic VSMCs from WT mice but not from KO mice. Phenylephrine induced a dose-dependent contraction of WT aortic segments, and this response was inhibited by catalase. Moreover, H(2)O(2) itself evoked Ca(2+) influx and inward currents in A7r5 cells, and these responses were significantly attenuated by either inhibition of TRPC6 or blocking vesicle trafficking. H(2)O(2) also induced inward currents in primary VSMCs from WT but not from TRPC6 KO mice. Additionally, H(2)O(2) stimulated a dose-dependent constriction of the aortas from WT mice but not from the vessels of KO mice. Furthermore, TIRFM showed that H(2)O(2) triggered membrane trafficking of TRPC6 in A7r5 cells. These results suggest a new signaling pathway of ROS-TRPC6 in controlling vessel contraction by vasoconstrictors.


Asunto(s)
Células Musculares/metabolismo , Músculo Liso Vascular/fisiología , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Aorta Torácica , Humanos , Peróxido de Hidrógeno/farmacología , Ratones , Ratones Noqueados , Miocitos del Músculo Liso , Transporte de Proteínas/efectos de los fármacos , Especies Reactivas de Oxígeno/farmacología , Canal Catiónico TRPC6 , Rigidez Vascular/efectos de los fármacos , Vasoconstrictores/farmacología
3.
J Mol Cell Cardiol ; 51(2): 177-86, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21616078

RESUMEN

Myocardial contractile dysfunction is a major consequence of septic shock, which is mainly mediated by nuclear factor-kappa B (NF-кB)-dependent production of inflammatory mediators in the heart. A novel zinc-finger protein, MCP-1-induced protein (MCPIP), is thought to have NF-кB inhibitory activity in certain cell cultures, but its pathophysiological consequence in vivo remains undefined. This study aims to clarify whether the anti-inflammatory potency of MCPIP contribute to amelioration of septic myocardial inflammation and dysfunction in vivo. Transgenic mice (TG) with cardiac-specific expression of MCPIP and their littermate wild-type (WT) controls were challenged with Escherichia coli LPS (10mg/kg ip) and myocardial function was assessed 18 h later using echocardiography. LPS administration markedly deteriorated myocardial contractile function evidenced by reduction of the percentage of left ventricular fractional shortening, which was significantly attenuated by myocardial expression of MCPIP. MCPIP TG mice exhibited a markedly reduced myocardial inflammatory cytokines, less of iNOS expression and peroxynitrite formation, decreased caspase-3/7 activities and apoptotic cell death compared with LPS-treated WT mice. Activation of cardiac NF-кB observed in LPS-challenged WT mice was suppressed by the presence of MCPIP, as evidenced by decreased phosphorylation of IкB kinase (IKKα/ß), reduced degradation of the cytosolic IкBα, and decreased nuclear translocation of NF-кB p65 subunit and its target DNA-binding activity. These results suggest that MCPIP has therapeutic values to protect heart from inflammatory pathologies, possibly through inhibition of IкB kinase complex, leading to blockade of NF-кB activation, and subsequently, attenuation of the proinflammatory state and nitrosative stress in the myocardium.


Asunto(s)
Cardiomiopatías/inducido químicamente , Activación Enzimática , Corazón/fisiopatología , Quinasa I-kappa B/antagonistas & inhibidores , Miocardio/enzimología , FN-kappa B/metabolismo , Ribonucleasas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Inflamación/genética , Inflamación/metabolismo , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Transgénicos , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Ácido Peroxinitroso/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Ribonucleasas/genética , Sepsis/inducido químicamente , Sepsis/genética , Sepsis/metabolismo
4.
Am J Physiol Cell Physiol ; 301(2): C304-15, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21525431

RESUMEN

The present study was performed to investigate the underlying mechanism, particularly the roles of reactive oxygen species (ROS) and protein kinase C (PKC), in the diabetes-induced canonical transient receptor potential 6 (TRPC6) downregulation. We found that high glucose (HG) significantly reduced TRPC6 protein expression in cultured mesangial cells (MCs). TRPC6 protein was also significantly reduced in the glomeruli but not in the heart or aorta isolated from streptozotocin-induced diabetic rats. In the cultured MCs, H(2)O(2) suppressed TRPC6 protein expression in a dose- and time-dependent manner, which emulated the HG effect. Catalase as well as superoxide dismutase were able to prevent the inhibitory effect of HG on TRPC6. The antioxidant effect observed in cultured cells was also observed in diabetic rats treated with tempol for 2 wk, which exhibited a preservation of TRPC6 in the glomeruli. Specific knockdown of Nox4, a component of NADPH oxidase, increased TRPC6 protein expression. Furthermore, the PKC activator phorbol 12-myristate 13-acetate (PMA), but not its analog 4α-phorbol 12, 13-didecanoate (4α-PDD), suppressed TRPC6 expression, and this PMA effect was not affected by catalase. Moreover, Gö6976, but not LY333531, attenuated the negative effect of HG on TRPC6 expression. Gö6976 also inhibited H(2)O(2) effect on TRPC6. Furthermore, either knockdown of TRPC6 or HG treatment significantly decreased ANG II-stimulated MC contraction, and the HG-impaired MC contraction was rescued by overexpression of TRPC6. These results suggest that hyperglycemia in diabetes downregulated TRPC6 protein expression in MCs through a NADPH oxidase Nox4-ROS-PKC pathway, proving a mechanism for impaired MC contraction in diabetes.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Nefropatías Diabéticas/etiología , Glucosa/metabolismo , Células Mesangiales/enzimología , Estrés Oxidativo , Proteína Quinasa C/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Canales Catiónicos TRPC/metabolismo , Análisis de Varianza , Angiotensina II/metabolismo , Animales , Antioxidantes/farmacología , Glucemia/metabolismo , Forma de la Célula , Células Cultivadas , Diabetes Mellitus Experimental/enzimología , Diabetes Mellitus Experimental/patología , Nefropatías Diabéticas/enzimología , Nefropatías Diabéticas/patología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Activación Enzimática , Activadores de Enzimas/farmacología , Humanos , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/patología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Oxidantes/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/metabolismo , Transducción de Señal , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6 , Factores de Tiempo
5.
J Biol Chem ; 285(30): 23466-76, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20501650

RESUMEN

This study examined the effect of H(2)O(2) on the TRPC6 channel and its underlying mechanisms using a TRPC6 heterologous expression system. In TRPC6-expressing HEK293T cells, H(2)O(2) significantly stimulated Ca(2+) entry in a dose-dependent manner. Electrophysiological experiments showed that H(2)O(2) significantly increased TRPC6 channel open probability and whole-cell currents. H(2)O(2) also evoked a robust inward current in A7r5 vascular smooth muscle cells, which was nearly abolished by knockdown of TRPC6 using a small interfering RNA. Catalase substantially attenuated arginine vasopressin (AVP)-induced Ca(2+) entry in cells co-transfected with TRPC6 and AVP V1 receptor. N-Ethylmaleimide and thimerosal were able to simulate the H(2)O(2) response. Dithiothreitol or glutathione-reduced ethyl ester significantly antagonized the response. Furthermore, both N-ethylmaleimide- and H(2)O(2)-induced TRPC6 activations were only observed in the cell-attached patches but not in the inside-out patches. Moreover, 1-oleoyl-2-acetyl-sn-glycerol effect on TRPC6 was significantly greater in the presence of H(2)O(2). Biotinylation assays revealed a significant increase in cell surface TRPC6 in response to H(2)O(2). Similarly, in cells transfected with TRPC6-EGFP, confocal microscopy showed a significant increase in fluorescence intensity in the region of the cell membrane and adjacent to the membrane. AVP also increased the fluorescence intensity on the surface of the cells co-transfected with TRPC6-EGFP and V1 receptor, and this response was inhibited by catalase. These data indicate that H(2)O(2) activates TRPC6 channels via modification of thiol groups of intracellular proteins. This cysteine oxidation-dependent pathway not only stimulates the TRPC6 channel by itself but also sensitizes the channels to diacylglycerol and promotes TRPC6 trafficking to the cell surface.


Asunto(s)
Canales Catiónicos TRPC/metabolismo , Animales , Calcio/metabolismo , Bovinos , Línea Celular , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Diglicéridos/metabolismo , Conductividad Eléctrica , Humanos , Peróxido de Hidrógeno/farmacología , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Oxidación-Reducción , Transporte de Proteínas/efectos de los fármacos , Ratas , Compuestos de Sulfhidrilo/metabolismo , Canales Catiónicos TRPC/química
6.
Exp Biol Med (Maywood) ; 234(6): 673-82, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19307462

RESUMEN

Although Orai1 protein was recently identified as the component of CRAC channels in hematopoietic cells, store-operated channels (SOC) in other cell types may have a different molecular entity. Also, the activation mechanism of SOC remains unclear, in general. In the present study, we tested the hypothesis that TRPC1 and TRPC4 proteins were functional subunits of SOC in glomerular mesangial cells (MCs) and that STIM1 was required for the channel activation through interaction with the TRPC proteins. In cultured human MCs, cell-attached patch clamp and fura-2 fluorescence measurements showed that single knockdown of either TRPC1 or TRPC4 significantly attenuated thapsigargin-induced membrane currents and Ca2+ entry as well as Ang II-induced channel activity. Double knockdown of both TRPCs resulted in a comparable inhibition on store-operated Ca2+ entry with single knockdown of either TRPC. Consistent with our previous report, co-immunoprecipitation showed a physical interaction between TRPC1 and TRPC4. Furthermore, we found that knockdown of STIM1 using RNAi significantly reduced the thapsigargin-stimulated membrane currents. Co-immunoprecipitation showed that STIM1 interacted with TRPC4, but not TRPC1. In addition, simultaneous inhibition of STIM1 and TRPC1 resulted in a comparable effect on SOC with single inhibition of either one of them. Taken together, we conclude that in glomerular mesangial cells, the TRPC1/TRPC4 complexes constitute the functional subunits of SOC and that the interaction between STIM1 and TRPC4 may be the mechanism for the activation of the channels.


Asunto(s)
Calcio/metabolismo , Mesangio Glomerular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canales Catiónicos TRPC/metabolismo , Angiotensina II/farmacología , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Humanos , Transporte Iónico/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Molécula de Interacción Estromal 1 , Canales Catiónicos TRPC/genética , Tapsigargina/farmacología , Vasoconstrictores/farmacología
7.
Circ Res ; 98(9): 1177-85, 2006 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-16574901

RESUMEN

Monocyte chemoattractant protein-1 (MCP-1; CCL2)-mediated inflammation plays a critical role in the development of ischemic heart disease (IHD). However, the gene expression changes caused by signal transduction, triggered by MCP-1 binding to its receptor CCR2, and their possible role in the development of IHD are not understood. We present evidence that MCP-1 binding to CCR2 induces a novel transcription factor (MCP-induced protein [MCPIP]) that causes cell death. Gene microarray analysis showed that when expressed in hiuman embryonic kidney 293 cells, MCPIP induced apoptotic gene families before causing cell death. Mutagenesis studies showed that the structural features required for transcription factor-like activity were also required for causing cell death. Activation of caspase-3 was detected after MCPIP transfection and Z-VAD-fmk partially inhibited cell death. Cardiomyocyte-targeted expression of MCP-1 in mice caused death by heart failure at 6 months of age. MCPIP expression increased in parallel with the development of ventricular dysfunction. In situ hybridization showed the presence of MCPIP transcripts in the cardiomyocytes and immunohistochemistry showed that MCPIP was associated with the cardiomyocyte nuclei of apoptotic cardiomyocytes. CCR2 expression in cardiomyocytes increased with the development of IHD. MCPIP production induced by MCP-1 binding to CCR2 in the cardiomyocytes is probably involved in the development of IHD in this murine model. MCPIP transcript levels were much higher in the explanted human hearts with IHD than with nonischemic heart disease. These results provide a molecular insight into how chronic inflammation and exposure to MCP-1 contributes to heart failure and suggest that MCPIP could be a potential target for therapeutic intervention.


Asunto(s)
Apoptosis , Quimiocina CCL2/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Quimiocina/metabolismo , Factores de Transcripción/biosíntesis , Disfunción Ventricular/etiología , Animales , Muerte Celular , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Isquemia Miocárdica/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores CCR2 , Receptores de Quimiocina/biosíntesis , Ribonucleasas , Distribución Tisular , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Disfunción Ventricular/metabolismo
8.
Exp Biol Med (Maywood) ; 237(2): 111-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22282397

RESUMEN

Canonical transient receptor potential (TRPC) channel proteins have been identified as downstream molecules in a G protein-coupled receptor signaling pathway and are involved in a variety of cell functions due to their ability to regulate intracellular calcium signaling. TRPC channel physiology has been an increasingly interesting and relevant topic over the last decade, and the outcomes from various studies have advanced our understanding of TRPC function in the normal state. Recently, attention has turned to whether or not TRPC proteins are implicated in diseases. Emerging evidence suggests a significant contribution of several isoforms of TRPC proteins to cardiovascular as well as renal diseases. This review focuses on the implication of TRPC proteins as they pertain to diabetes. We summarize the recent findings by other investigators as well as ourselves and additionally discuss the important role of TRPC proteins in the development of various diabetic complications, such as diabetic nephropathy and diabetic vasculopathy. The underlying mechanisms which contribute to these complications are also outlined. Lastly, we elaborate on the role of TRPC proteins as a potential therapeutic target for treating diabetes-associated diseases.


Asunto(s)
Diabetes Mellitus/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Canales Catiónicos TRPC/metabolismo , Animales , Aterosclerosis/metabolismo , Calcio/química , Calcio/metabolismo , Señalización del Calcio , Complicaciones de la Diabetes/metabolismo , Complicaciones de la Diabetes/patología , Glucosa/metabolismo , Humanos , Modelos Biológicos , Isoformas de Proteínas/química , Especies Reactivas de Oxígeno , Transducción de Señal , Canales Catiónicos TRPC/fisiología
9.
Am J Physiol Renal Physiol ; 294(4): F909-18, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18256307

RESUMEN

Ca(+) influx across the plasma membrane is a major component of mesangial cell (MC) response to vasoconstrictors. Polycystin 2 (PC2), the protein product of the gene mutated in type 2 autosomal dominant polycystic kidney disease, has been shown to function as a nonselective cation channel in a variety of cell types. The present study was performed to test the hypothesis that PC2 and its binding partners constitute a Ca(2+)-permeable channel and contribute to ANG II-induced Ca(2+) signaling in MCs. Western blot and immunocytochemistry showed PC2 expression in cultured human MCs. The existence of PC2 in MCs was further confirmed by immunohistochemsitry in rat kidney sections. Coimmunoprecipitation displayed a selective interaction of PC2 with canonical transient receptor potential (TRPC) proteins TRPC1 and TRPC4. Cell-attached patch-clamp experiments revealed that ANG II-induced membrane currents were enhanced by overexpression of pkd2 but significantly inhibited by knock down of pkd2, 30 microM Gd(3+) (a PC2 channel blocker), and dominant-negative pkd2 mutant (pkd2-D511V). Corresponding to the increase in channel currents, ANG II stimulation increased expression of PC2 on the cell surface of MCs and interaction with TRPC1 and TRPC4. Furthermore, ANG II-induced MC contraction was significantly reduced in pkd2-knocked down MCs. These data suggest that PC2 selectively assembles with TRPC1 and TRPC4 to form channel complexes mediating ANG II-induced Ca(2+) responses in MCs.


Asunto(s)
Angiotensina II/farmacología , Calcio/fisiología , Mesangio Glomerular/fisiología , Transducción de Señal/fisiología , Canales Catiónicos TRPP/metabolismo , Biotinilación , Células Cultivadas , Mesangio Glomerular/citología , Mesangio Glomerular/efectos de los fármacos , Humanos , Immunoblotting , Microscopía Fluorescente , Técnicas de Placa-Clamp , Transducción de Señal/efectos de los fármacos
10.
Artículo en Inglés | MEDLINE | ID: mdl-26023301

RESUMEN

Inositol 1, 4, 5-trisphosphate (IP3) receptor associated cyclic GMP (cGMP) kinase substrate (IRAG, also known as Mrv1) is a type-2 integral membrane endoplasmic reticulum (ER) protein, which interacts with IP3 Receptor type 1 (IP3R1), cGMP kinase I-ß (cGKI ß) and other associated proteins. It plays a key role in NO, cGMP, and cGKI ß mediated inhibition of IP3R1 activity and thus relaxes smooth muscle tone and inhibits platelet aggregation. As a scaffolding protein Mrv1 maintains the conformation of a heteroprotein complex containing cGKI ß, IP3R1 and other proteins and enables efficient activity of cGKI ß within the complex. Increased expression of Mrv1 or IRAG in the absence of tumor related transcription factor in pancreatic cancer cells suggest that it might be involved in tumorigenesis. Downregulation of Mrv1 during megakaryocyte maturation indicates that it is involved in cell growth and differentiation.

11.
Am J Physiol Renal Physiol ; 293(4): F1381-90, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17699555

RESUMEN

The present study was performed to investigate whether transient receptor potential (TRPC)6 participated in Ca(2+) signaling of glomerular mesangial cells (MCs) and expression of this protein was altered in diabetes. Western blots and real-time PCR were used to evaluate the expression level of TRPC6 protein and mRNA, respectively. Cell-attached patch-clamp and fura-2 fluorescence measurements were utilized to assess angiotensin II (ANG II)-stimulated membrane currents and Ca(2+) responses in MCs. In cultured human MCs, high glucose significantly reduced expression of TRPC6 protein, but there was no effect on either TRPC1 or TRPC3. The high glucose-induced effect on TRPC6 was time and dose dependent with the maximum effect observed on day 7 and at 30 mM glucose, respectively. In glomeruli isolated from streptozotocin-induced diabetic rats, TRPC6, but not TRPC1, was markedly reduced compared with the glomeruli of control rats. Furthermore, TRPC6 mRNA in MCs was also significantly decreased by high glucose as early as 1 day after treatment with maximal reduction on day 4. Patch-clamp experiments showed that ANG II-stimulated membrane currents in MCs were significantly attenuated or enhanced by knockdown or overexpression of TRPC6, respectively. Fura-2 fluorescence measurements revealed that the ANG II-induced Ca(2+) influxes were markedly inhibited in MCs with TRPC6 knockdown, reminiscent of the impaired Ca(2+) entry in response to ANG II in high glucose-treated MCs. These results suggest that the TRPC6 protein expression in MCs was downregulated by high glucose and the deficiency of TRPC6 protein might contribute to the impaired Ca(2+) signaling of MCs seen in diabetes.


Asunto(s)
Señalización del Calcio/fisiología , Diabetes Mellitus Experimental/metabolismo , Glucosa/fisiología , Células Mesangiales/metabolismo , Canales Catiónicos TRPC/metabolismo , Angiotensina II/farmacología , Animales , Canales de Calcio/efectos de los fármacos , Canales de Calcio/fisiología , Células Cultivadas , Diabetes Mellitus Experimental/patología , Regulación hacia Abajo , Humanos , Masculino , Células Mesangiales/patología , Técnicas de Placa-Clamp , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Estreptozocina , Canales Catiónicos TRPC/genética , Canal Catiónico TRPC6
12.
J Am Soc Nephrol ; 18(5): 1437-45, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17389736

RESUMEN

Contractility of mesangial cells (MC) is tightly controlled by [Ca(2+)](i). Ca(2+) influx across the plasma membrane constitutes a major component of mesangial responses to vasoconstrictors. Canonical transient receptor potential 1 (TRPC1) is a Ca(2+)-permeable cation channel in a variety of cell types. This study was performed to investigate whether TRPC1 takes part in vasoconstrictor-induced mesangial contraction by mediating Ca(2+) entry. It was found that angiotensin II (AngII) evoked remarkable contraction of the cultured MC. Downregulation of TRPC1 using RNA interference significantly attenuated the contractile response. Infusion of AngII or endothelin-1 in rats caused a decrease in GFR. The GFR decline was significantly reduced by infusion of TRPC1 antibody that targets an extracellular domain in the pore region of TRPC1 channel. However, the treatment of TRPC1 antibody did not affect the AngII-induced vasopressing effect. Electrophysiologic experiments revealed that functional or biologic inhibition of TRPC1 significantly depressed AngII-induced channel activation. Fura-2 fluorescence-indicated that Ca(2+) entry in response to AngII stimulation was also dramatically inhibited by TRPC1 antibody and TRPC1-specific RNA interference. These results suggest that TRPC1 plays an important role in controlling contractile function of MC. Mediation of Ca(2+) entry might be the underlying mechanism for the TRPC1-associated MC contraction.


Asunto(s)
Células Mesangiales/fisiología , Contracción Muscular/fisiología , Canales Catiónicos TRPC/fisiología , Angiotensina II/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Tasa de Filtración Glomerular/fisiología , Humanos , Masculino , Potenciales de la Membrana/efectos de los fármacos , Contracción Muscular/efectos de los fármacos , Concentración Osmolar , Ratas , Ratas Sprague-Dawley , Equilibrio Hidroelectrolítico/fisiología
13.
Stem Cells ; 22(7): 1305-20, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15579648

RESUMEN

Muscle progenitor cells (satellite cells) function in the maintenance and repair of adult skeletal muscle. Side population (SP) cells are enriched in repopulating activity and also reside in adult skeletal muscle. In this study, we observed that Abcg2 is a determinant of the SP cell phenotype. Using reverse transcription polymerase chain reaction and immunohistochemical techniques, we localized Abcg2-expressing cells in the interstitium and in close approximation to the vasculature of adult skeletal muscle. Muscle SP cells are able to differentiate into myotubes and increase in number after cardiotoxin-induced muscle injury. Similar to myogenic progenitor cells, muscle SP cells express Foxk1 and are decreased in number in Foxk1 mutant skeletal muscle. Using emerging technologies, we examine the molecular signature of muscle SP cells from normal, injured, and Foxk1 mutant skeletal muscle to define common and distinct molecular programs. We propose that muscle SP cells are progenitor cells that participate in repair and regeneration of adult skeletal muscle.


Asunto(s)
Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/citología , Células Satélite del Músculo Esquelético/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/fisiología , Animales , Diferenciación Celular , Separación Celular , Proteínas Cardiotóxicas de Elápidos/farmacología , Citometría de Flujo , Factores de Transcripción Forkhead , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculos/patología , Distrofias Musculares/patología , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/genética , Hibridación de Ácido Nucleico , Fenotipo , ARN/metabolismo , Regeneración , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Factores de Tiempo , Factores de Transcripción/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA