Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
J Mol Cell Cardiol ; 103: 1-10, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28017639

RESUMEN

Sphingosine-1-phosphate (S1P), a bioactive lysophospholipid, is generated and released at sites of tissue injury in the heart and can act on S1P1, S1P2, and S1P3 receptor subtypes to affect cardiovascular responses. We established that S1P causes little phosphoinositide hydrolysis and does not induce hypertrophy indicating that it does not cause receptor coupling to Gq. We previously demonstrated that S1P confers cardioprotection against ischemia/reperfusion by activating RhoA and its downstream effector PKD. The S1P receptor subtypes and G proteins that regulate RhoA activation and downstream responses in the heart have not been determined. Using siRNA or pertussis toxin to inhibit different G proteins in NRVMs we established that S1P regulates RhoA activation through Gα13 but not Gα12, Gαq, or Gαi. Knockdown of the three major S1P receptors using siRNA demonstrated a requirement for S1P3 in RhoA activation and subsequent phosphorylation of PKD, and this was confirmed in studies using isolated hearts from S1P3 knockout (KO) mice. S1P treatment reduced infarct size induced by ischemia/reperfusion in Langendorff perfused wild-type (WT) hearts and this protection was abolished in the S1P3 KO mouse heart. CYM-51736, an S1P3-specific agonist, also decreased infarct size after ischemia/reperfusion to a degree similar to that achieved by S1P. The finding that S1P3 receptor- and Gα13-mediated RhoA activation is responsible for protection against ischemia/reperfusion suggests that selective targeting of S1P3 receptors could provide therapeutic benefits in ischemic heart disease.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proproteína Convertasas/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Serina Endopeptidasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Cardiomegalia/patología , Lisofosfolípidos/metabolismo , Masculino , Ratones , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Unión Proteica , Ratas , Transducción de Señal , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Canales Catiónicos TRPP/metabolismo
2.
J Mol Cell Cardiol ; 75: 152-61, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25106095

RESUMEN

Activation of RhoA, a low molecular-weight G-protein, plays an important role in protecting the heart against ischemic stress. Studies using non-cardiac cells demonstrate that the expression and subsequent secretion of the matricellular protein CCN1 is induced by GPCR agonists that activate RhoA. In this study we determined whether and how CCN1 is induced by GPCR agonists in cardiomyocytes and examined the role of CCN1 in ischemic cardioprotection in cardiomyocytes and the isolated perfused heart. In neonatal rat ventricular myocytes (NRVMs), sphingosine 1-phosphate (S1P), lysophosphatidic acid (LPA) and endothelin-1 induced robust increases in CCN1 expression while phenylephrine, isoproterenol and carbachol had little or no effect. The ability of agonists to activate the small G-protein RhoA correlated with their ability to induce CCN1. CCN1 induction by S1P was blocked when RhoA function was inhibited with C3 exoenzyme or a pharmacological RhoA inhibitor. Conversely overexpression of RhoA was sufficient to induce CCN1 expression. To delineate the signals downstream of RhoA we tested the role of MRTF-A (MKL1), a co-activator of SRF, in S1P-mediated CCN1 expression. S1P increased the nuclear accumulation of MRTF-A and this was inhibited by the functional inactivation of RhoA. In addition, pharmacological inhibitors of MRTF-A or knockdown of MRTF-A significantly diminished S1P-mediated CCN1 expression, indicating a requirement for RhoA/MRTF-A signaling. We also present data indicating that CCN1 is secreted following agonist treatment and RhoA activation, and binds to cells where it can serve an autocrine function. To determine the functional significance of CCN1 expression and signaling, simulated ischemia/reperfusion (sI/R)-induced apoptosis was assessed in NRVMs. The ability of S1P to protect against sI/R was significantly reduced by the inhibition of RhoA, ROCK or MRTF-A or by CCN1 knockdown. We also demonstrate that ischemia/reperfusion induces CCN1 expression in the isolated perfused heart and that this functions as a cardioprotective mechanism, evidenced by the significant increase in infarct development in response to I/R in the cardiac specific CCN1 KO relative to control mice. Our findings implicate CCN1 as a mediator of cardioprotection induced by GPCR agonists that activate RhoA/MRTF-A signaling.


Asunto(s)
Cardiotónicos/metabolismo , Proteína 61 Rica en Cisteína/metabolismo , Isquemia Miocárdica/metabolismo , Factores de Transcripción/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Animales Recién Nacidos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Ventrículos Cardíacos/citología , Técnicas In Vitro , Lisofosfolípidos/farmacología , Ratones Noqueados , Modelos Biológicos , Isquemia Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Unión Proteica/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Esfingosina/análogos & derivados , Esfingosina/farmacología
3.
Sci Signal ; 6(306): ra108, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24345679

RESUMEN

Activation of the small guanosine triphosphatase RhoA can promote cell survival in cultured cardiomyocytes and in the heart. We showed that the circulating lysophospholipid sphingosine 1-phosphate (S1P), a G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor (GPCR) agonist, signaled through RhoA and phospholipase Cε (PLCε) to increase the phosphorylation and activation of protein kinase D1 (PKD1). Genetic deletion of either PKD1 or its upstream regulator PLCε inhibited S1P-mediated cardioprotection against ischemia/reperfusion injury. Cardioprotection involved PKD1-mediated phosphorylation and inhibition of the cofilin phosphatase Slingshot 1L (SSH1L). Cofilin 2 translocates to mitochondria in response to oxidative stress or ischemia/reperfusion injury, and both S1P pretreatment and SSH1L knockdown attenuated translocation of cofilin 2 to mitochondria. Cofilin 2 associates with the proapoptotic protein Bax, and the mitochondrial translocation of Bax in response to oxidative stress was also attenuated by S1P treatment in isolated hearts or by knockdown of SSH1L or cofilin 2 in cardiomyocytes. Furthermore, SSH1L knockdown, like S1P treatment, increased cardiomyocyte survival and preserved mitochondrial integrity after oxidative stress. These findings reveal a pathway initiated by GPCR agonist-induced RhoA activation, in which PLCε signals to PKD1-mediated phosphorylation of cytoskeletal proteins to prevent the mitochondrial translocation and proapoptotic function of cofilin 2 and Bax and thereby promote cell survival.


Asunto(s)
Mitocondrias Cardíacas/metabolismo , Estrés Oxidativo , Fosfoinositido Fosfolipasa C/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Proteína Quinasa C/metabolismo , Transducción de Señal , Proteína de Unión al GTP rhoA/metabolismo , Animales , Cofilina 2/metabolismo , Peróxido de Hidrógeno/farmacología , Lisofosfolípidos/metabolismo , Ratones , Transporte de Proteínas , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Proteína X Asociada a bcl-2/metabolismo
4.
J Cardiovasc Transl Res ; 3(4): 330-43, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20559774

RESUMEN

The neonatal rat ventricular myocyte model of hypertrophy has provided tremendous insight with regard to signaling pathways regulating cardiac growth and gene expression. Many mediators thus discovered have been successfully extrapolated to the in vivo setting, as assessed using genetically engineered mice and physiological interventions. Studies in neonatal rat ventricular myocytes demonstrated a role for the small G-protein RhoA and its downstream effector kinase, Rho-associated coiled-coil containing protein kinase (ROCK), in agonist-mediated hypertrophy. Transgenic expression of RhoA in the heart does not phenocopy this response, however, nor does genetic deletion of ROCK prevent hypertrophy. Pharmacologic inhibition of ROCK has effects most consistent with roles for RhoA signaling in the development of heart failure or responses to ischemic damage. Whether signals elicited downstream of RhoA promote cell death or survival and are deleterious or salutary is, however, context and cell-type dependent. The concepts discussed above are reviewed, and the hypothesis that RhoA might protect cardiomyocytes from ischemia and other insults is presented. Novel RhoA targets including phospholipid regulated and regulating enzymes (Akt, PI kinases, phospholipase C, protein kinases C and D) and serum response element-mediated transcriptional responses are considered as possible pathways through which RhoA could affect cardiomyocyte survival.


Asunto(s)
Cardiomegalia/enzimología , Insuficiencia Cardíaca/enzimología , Corazón/fisiopatología , Miocardio/enzimología , Quinasas Asociadas a rho/metabolismo , Animales , Biomarcadores/metabolismo , Cardiomegalia/genética , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Ratones , Miocitos Cardíacos/enzimología , Ratas , Transducción de Señal , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/biosíntesis , Proteína de Unión al GTP rhoA/metabolismo
5.
Acta Pharmacol Sin ; 26(3): 265-78, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15715921

RESUMEN

In comparison to cation (K+, Na+, and Ca2+) channels, much less is currently known about the functional role of anion (Cl-) channels in cardiovascular physiology and pathophysiology. Over the past 15 years, various types of Cl- currents have been recorded in cardiac cells from different species including humans. All cardiac Cl- channels described to date may be encoded by five different Cl- channel genes: the PKA- and PKC-activated cystic fibrosis tansmembrane conductance regulator (CFTR), the volume-regulated ClC-2 and ClC-3, and the Ca2+-activated CLCA or Bestrophin. Recent studies using multiple approaches to examine the functional role of Cl- channels in the context of health and disease have demonstrated that Cl- channels might contribute to: 1) arrhythmogenesis in myocardial injury; 2) cardiac ischemic preconditioning; and 3) the adaptive remodeling of the heart during myocardial hypertrophy and heart failure. Therefore, anion channels represent very attractive novel targets for therapeutic approaches to the treatment of heart diseases. Recent evidence suggests that Cl- channels, like cation channels, might function as a multiprotein complex or functional module. In the post-genome era, the emergence of functional proteomics has necessitated a new paradigm shift to the structural and functional assessment of integrated Cl- channel multiprotein complexes in the heart, which could provide new insight into our understanding of the underlying mechanisms responsible for heart disease and protection.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Cardiomegalia/fisiopatología , Canales de Cloruro/fisiología , Precondicionamiento Isquémico , Animales , Arritmias Cardíacas/genética , Cardiomegalia/genética , Cardiomegalia/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/fisiología , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA