Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 72(15): 2779-92, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26091747

RESUMEN

Connexins and pannexins form connexons, pannexons and membrane channels, which are critically involved in many aspects of cardiovascular physiology. For that reason, a vast number of studies have addressed the role of connexins and pannexins in the arterial and venous systems as well as in the heart. Moreover, a role for connexins in lymphatics has recently also been suggested. This review provides an overview of the current knowledge regarding the involvement of connexins and pannexins in cardiovascular physiology.


Asunto(s)
Sistema Cardiovascular/metabolismo , Conexinas/metabolismo , Animales , Fenómenos Fisiológicos Cardiovasculares , Uniones Comunicantes/metabolismo , Uniones Comunicantes/fisiología , Humanos , Canales Iónicos/metabolismo
2.
J Biol Chem ; 288(43): 31139-53, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24022490

RESUMEN

Humans and other higher primates are unique among mammals in using complement receptor 1 (CR1, CD35) on red blood cells (RBC) to ligate complement-tagged inflammatory particles (immune complexes, apoptotic/necrotic debris, and microbes) in the circulation for quiet transport to the sinusoids of spleen and liver where resident macrophages remove the particles, but allow the RBC to return unharmed to the circulation. This process is called immune-adherence clearance. In this study we found using luminometric- and fluorescence-based methods that ligation of CR1 on human RBC promotes ATP release. Our data show that CR1-mediated ATP release does not depend on Ca(2+) or enzymes previously shown to mediate an increase in membrane deformability promoted by CR1 ligation. Furthermore, ATP release following CR1 ligation increases the mobility of the lipid fraction of RBC membranes, which in turn facilitates CR1 clustering, and thereby enhances the binding avidity of complement-opsonized particles to the RBC CR1. Finally, we have found that RBC-derived ATP has a stimulatory effect on phagocytosis of immune-adherent immune complexes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Eritrocitos/metabolismo , Recubrimiento Inmunológico , Receptores de Complemento 3b/metabolismo , Adenosina Trifosfato/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Eritrocitos/citología , Eritrocitos/inmunología , Femenino , Humanos , Masculino , Lípidos de la Membrana/inmunología , Lípidos de la Membrana/metabolismo , Fagocitosis/inmunología , Receptores de Complemento 3b/inmunología
3.
Circulation ; 125(3): 474-81, 2012 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-22179534

RESUMEN

BACKGROUND: An estimated 10% to 15% of sudden infant death syndrome (SIDS) cases may stem from channelopathy-mediated lethal arrhythmias. Loss of the GJA1-encoded gap junction channel protein connexin43 is known to underlie formation of lethal arrhythmias. GJA1 mutations have been associated with cardiac diseases, including atrial fibrillation. Therefore, GJA1 is a plausible candidate gene for premature sudden death. METHODS AND RESULTS: GJA1 open reading frame mutational analysis was performed with polymerase chain reaction, denaturing high-performance liquid chromatography, and direct DNA sequencing on DNA from 292 SIDS cases. Immunofluorescence and dual whole-cell patch-clamp studies were performed to determine the functionality of mutant gap junctions. Immunostaining for gap junction proteins was performed on SIDS-associated paraffin-embedded cardiac tissue. Two rare, novel missense mutations, E42K and S272P, were detected in 2 of 292 SIDS cases, a 2-month-old white boy and a 3-month-old white girl, respectively. Analysis of the E42K victim's parental DNA demonstrated a de novo mutation. Both mutations involved highly conserved residues and were absent in >1000 ethnically matched reference alleles. Immunofluorescence demonstrated no trafficking abnormalities for either mutation, and S272P demonstrated wild-type junctional conductance. However, junctional conductance measurements for the E42K mutation demonstrated a loss of function not rescued by wild type. Moreover, the E42K victim's cardiac tissue demonstrated a mosaic immunostaining pattern for connexin43 protein. CONCLUSIONS: This study provides the first molecular and functional evidence implicating a GJA1 mutation as a novel pathogenic substrate for SIDS. E42K-connexin43 demonstrated a trafficking-independent reduction in junctional coupling in vitro and a mosaic pattern of mutational DNA distribution in deceased cardiac tissue, suggesting a novel mechanism of connexin43-associated sudden death.


Asunto(s)
Conexina 43/genética , Uniones Comunicantes/patología , Uniones Comunicantes/fisiología , Mutación Missense , Muerte Súbita del Lactante/genética , Muerte Súbita del Lactante/patología , Adulto , Animales , Cadherinas/metabolismo , Estudios de Cohortes , Conexina 43/metabolismo , Desmoplaquinas/metabolismo , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Lactante , Masculino , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Transporte de Proteínas/genética , Ratas
4.
Arterioscler Thromb Vasc Biol ; 32(8): 1951-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22652601

RESUMEN

OBJECTIVE: Capillary network formation represents a specialized endothelial cell function and is a prerequisite to establish a continuous vessel lumen. Formation of endothelial cell connections that form the vascular structure is regulated, at least in part, at the transcriptional level. We report here that related transcription enhancer factor-1 (RTEF-1) plays an important role in vascular structure formation. METHODS AND RESULTS: Knockdown of RTEF-1 by small interfering RNA or blockage of RTEF-1 function by the transcription enhancer activators domain decreased endothelial connections in a Matrigel assay, whereas overexpression of RTEF-1 in endothelial cells resulted in a significant increase in cell connections and aggregation. In a model of oxygen-induced retinopathy, endothelial-specific RTEF-1 overexpressing mice had enhanced angiogenic sprouting and vascular structure remodeling, resulting in the formation of a denser and more highly interconnected superficial capillary plexus. Mechanistic studies revealed that RTEF-1 induced the expression of functional gap junction proteins including connexin 43, connexin 40, and connexin 37. Blocking connexin 43 function inhibited RTEF-1-induced endothelial cell connections and aggregation. CONCLUSIONS: These findings provide novel insights into the transcriptional control of endothelial function in the coordination of cell-cell connections.


Asunto(s)
Comunicación Celular , Conexinas/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Células Endoteliales/fisiología , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiología , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Animales , Agregación Celular , Células Cultivadas , Conexina 43/genética , Proteínas de Unión al ADN/genética , Humanos , Ratones , Proteínas Musculares/genética , Neovascularización Fisiológica , Factores de Transcripción de Dominio TEA , Factores de Transcripción/genética
5.
Am J Physiol Heart Circ Physiol ; 302(3): H790-800, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-22101526

RESUMEN

Following myocardial infarction (MI) inflammatory responses transform cardiac fibroblasts to myofibroblasts, which in vitro studies show form heterocellular gap junctions with cardiac myocytes via Connexin43 (Cx43). The ability to form heterocellular junctions in the intact heart and the impact of these junctions on propagation is unclear. We used a canine model of MI and characterized the distribution and quantity of myofibroblasts in surviving epicardial cells [epicardial border zone (EBZ)]. We found a significant increase in myofibroblasts within the EBZ and no gap junction plaques between myofibroblasts and myocytes. Because myofibroblasts produce IL-1ß, which downregulates Cx43, we asked whether myofibroblast proliferation causes loss of Cx43 near myofibroblast clusters. In vitro studies showed that IL-1ß caused loss of Cx43 and reduced coupling. Western blot showed a significant increase of IL-1ß in the EBZ, and immunohistochemistry showed a loss of Cx43 in regions of myofibroblasts in the intact heart. Additionally, dye studies in intact heart showed no coupling between myocytes and myofibroblasts. To quantify the effect of myofibroblasts on propagation we used a two-dimensional subcellular computer model of the EBZ, which showed that heterogeneities in myofibroblast density lead to conduction abnormalities. In conclusion, an increase of myofibroblasts in the infarcted heart causes heterogeneous Cx43 levels, possibly as a result of the release of IL-1ß and decreased cell-cell communication, which leads to conduction abnormalities following MI.


Asunto(s)
Comunicación Celular/fisiología , Conexina 43/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Cicatrización de Heridas/fisiología , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patología , Línea Celular , Simulación por Computador , Perros , Colorantes Fluorescentes/farmacocinética , Uniones Comunicantes/fisiología , Interleucina-1beta/metabolismo , Isoquinolinas/farmacocinética , Riñón/citología , Modelos Cardiovasculares , Infarto del Miocardio/patología , Miocitos Cardíacos/citología , Miofibroblastos/citología , Comunicación Paracrina/fisiología , Ratas
6.
Am J Physiol Heart Circ Physiol ; 303(10): H1208-18, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22982782

RESUMEN

Fibrosis following myocardial infarction is associated with increases in arrhythmias and sudden cardiac death. Initial steps in the development of fibrosis are not clear; however, it is likely that cardiac fibroblasts play an important role. In immune cells, ATP release from pannexin 1 (Panx1) channels acts as a paracrine signal initiating activation of innate immunity. ATP has been shown in noncardiac systems to initiate fibroblast activation. Therefore, we propose that ATP release through Panx1 channels and subsequent fibroblast activation in the heart drives the development of fibrosis in the heart following myocardial infarction. We identified for the first time that Panx1 is localized within sarcolemmal membranes of canine cardiac myocytes where it directly interacts with the postsynaptic density 95/Drosophila disk large/zonula occludens-1-containing scaffolding protein synapse-associated protein 97 via its carboxyl terminal domain (amino acids 300-357). Induced ischemia rapidly increased glycosylation of Panx1, resulting in increased trafficking to the plasma membrane as well as increased interaction with synapse-associated protein 97. Cellular stress enhanced ATP release from myocyte Panx1 channels, which, in turn, causes fibroblast transformation to the activated myofibroblast phenotype via activation of the MAPK and p53 pathways, both of which are involved in the development of cardiac fibrosis. ATP release through Panx1 channels in cardiac myocytes during ischemia may be an early paracrine event leading to profibrotic responses to ischemic cardiac injury.


Asunto(s)
Adenosina Trifosfato/metabolismo , Conexinas/metabolismo , Fibroblastos/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Comunicación Paracrina , Animales , Membrana Celular/metabolismo , Técnicas de Cocultivo , Conexinas/genética , Modelos Animales de Enfermedad , Perros , Fibroblastos/patología , Fibrosis , Glicosilación , Células de Riñón Canino Madin Darby , Ratones , Infarto del Miocardio/genética , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proteínas del Tejido Nervioso/genética , Fenotipo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Sarcolema/metabolismo , Transducción de Señal , Factores de Tiempo , Regulación hacia Arriba
7.
Circ Res ; 104(9): 1103-12, 2009 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-19342602

RESUMEN

Lateralization of the ventricular gap junction protein connexin 43 (Cx43) occurs in epicardial border zone myocytes following myocardial infarction (MI) and is arrhythmogenic. Alterations in Cx43 protein partners have been hypothesized to play a role in lateralization although mechanisms by which this occurs are unknown. To examine potential mechanisms we did nuclear magnetic resonance, yeast 2-hybrid, and surface plasmon resonance studies and found that the SH3 domain of the tyrosine kinase c-Src binds to the Cx43 scaffolding protein zonula occludens-1 (ZO-1) with a higher affinity than does Cx43. This suggests c-Src outcompetes Cx43 for binding to ZO-1, thus acting as a chaperone for ZO-1 and causing unhooking from Cx43. To determine whether c-Src/ZO-1 interactions affect Cx43 lateralization within the epicardial border zone, we performed Western blot, immunoprecipitation, and immunolocalization for active c-Src (p-cSrc) post-MI using a canine model of coronary occlusion. We found that post-MI p-cSrc interacts with ZO-1 as Cx43 begins to decrease its interaction with ZO-1 and undergo initial loss of intercalated disk localization. This indicates that the molecular mechanisms by which Cx43 is lost from the intercalated disk following MI includes an interaction of p-cSrc with ZO-1 and subsequent loss of scaffolding of Cx43 leaving Cx43 free to diffuse in myocyte membranes from areas of high Cx43, as at the intercalated disk, to regions of lower Cx43 content, the lateral myocyte membrane. Therefore shifts in Cx43 protein partners may underlie, in part, arrhythmogenesis in the post-MI heart.


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Proteínas de la Membrana/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Pericardio/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Secuencia de Aminoácidos , Animales , Unión Competitiva , Conexina 43/química , Modelos Animales de Enfermedad , Perros , Uniones Comunicantes/enzimología , Uniones Comunicantes/patología , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , Dominios PDZ , Pericardio/enzimología , Pericardio/patología , Fosforilación , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Transporte de Proteínas , Proteínas Proto-Oncogénicas pp60(c-src)/química , Resonancia por Plasmón de Superficie , Técnicas del Sistema de Dos Híbridos , Proteína de la Zonula Occludens-1 , Dominios Homologos src
8.
J Cardiovasc Pharmacol ; 57(4): 373-5, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21346592

RESUMEN

Fibroblasts and their activated phenotype known as myofibroblasts are nonexcitable cells found in all organs of the body. In the heart, fibroblasts, along with the endothelial and smooth muscle cells of the blood vessels, make up approximately 30% of tissue mass. In vitro, myofibroblasts cocultured with cardiac myocytes can propagate electrical signals down cellular strands indicating that under these conditions myofibroblasts are capable of depolarizing enough to maintain electrical propagation. This has obvious implications for cardiac biology if heterocellular coupling between fibroblasts and myocytes were to occur in the intact heart either under normal conditions or during cellular stress. The purpose of this review series is to highlight the newest information on cardiac fibroblasts and myofibroblasts and to review the data on their interactions with cardiac myocytes.


Asunto(s)
Fibroblastos/metabolismo , Miocitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Fibrosis , Humanos , Miocardio/patología
9.
J Cardiovasc Pharmacol ; 57(4): 376-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21297493

RESUMEN

Cardiac myocytes, although large enough to make up most of the heart volume, are only a minority of cells within the heart with fibroblasts and blood vessel components (endothelial and smooth muscle cells) making up the remainder of the heart. In recent years, there has been increasing interest in the nonmyocyte population within the heart. This is attributable, in part, to our increasing understanding of the biology of the nonmyocyte cell types and additionally it is the result of our awakening realization that these cells are not static but rather that they are dynamic in nature indicating that they play a more active role in cardiac function than previously imagined. Studies now show that fibroblasts are involved in formation of the extracellular matrix and they control the size of the extracellular matrix. Additionally, they participate in the repair process by differentiating into myofibroblasts, which are cells involved in the inflammatory response to injury. Myofibroblasts migrate to the sites of injury where they produce cytokines, thus enhancing the inflammatory response. This review discusses both structural and functional differences between the two cell types and examines the different roles of these two different cell types in the heart.


Asunto(s)
Fibroblastos/metabolismo , Miocardio/citología , Miofibroblastos/metabolismo , Transdiferenciación Celular , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Humanos , Inflamación/patología , Miocardio/patología , Miocitos Cardíacos/metabolismo
10.
Mol Ther Methods Clin Dev ; 21: 369-381, 2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-33898634

RESUMEN

Duchenne muscular dystrophy is characterized by structural degeneration of muscle, which is exacerbated by localized functional ischemia due to loss of nitric oxide synthase-induced vasodilation. Treatment strategies aimed at increasing vascular perfusion have been proposed. Toward this end, we have developed monoclonal antibodies (mAbs) that bind to the vascular endothelial growth factor (VEGF) receptor VEGFR-1 (Flt-1) and its soluble splice variant isoform (sFlt-1) leading to increased levels of free VEGF and proangiogenic signaling. The lead chimeric mAb, 21B3, had high affinity and specificity for both human and mouse sFlt-1 and inhibited VEGF binding to sFlt-1 in a competitive manner. Proof-of-concept studies in the mdx mouse model of Duchenne muscular dystrophy showed that intravenous administration of 21B3 led to elevated VEGF levels, increased vascularization and blood flow to muscles, and decreased fibrosis after 6-12 weeks of treatment. Greater muscle strength was also observed after 4 weeks of treatment. A humanized form of the mAb, 27H6, was engineered and demonstrated a comparable pharmacologic effect. Overall, administration of anti-Flt-1 mAbs in mdx mice inhibited the VEGF:Flt-1 interaction, promoted angiogenesis, and improved muscle function. These studies suggest a potential therapeutic benefit of Flt-1 inhibition for patients with Duchenne muscular dystrophy.

11.
Circulation ; 119(1): 19-27, 2009 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-19103989

RESUMEN

BACKGROUND: In depolarized myocardial infarct epicardial border zones, the cardiac sodium channel (SCN5A) is largely inactivated, contributing to low action potential upstroke velocity (V(max)), slow conduction, and reentry. We hypothesized that a fast inward current such as the skeletal muscle sodium channel (SkM1) operating more effectively at depolarized membrane potentials might restore fast conduction in epicardial border zones and be antiarrhythmic. METHODS AND RESULTS: Computer simulations were done with a modified Hund-Rudy model. Canine myocardial infarcts were created by coronary ligation. Adenovirus expressing SkM1 and green fluorescent protein or green fluorescent protein alone (sham) was injected into epicardial border zones. After 5 to 7 days, dogs were studied with epicardial mapping, programmed premature stimulation in vivo, and cellular electrophysiology in vitro. Infarct size was determined, and tissues were immunostained for SkM1 and green fluorescent protein. In the computational model, modest SkM1 expression preserved fast conduction at potentials as positive as -60 mV; overexpression of SCN5A did not. In vivo epicardial border zone electrograms were broad and fragmented in shams (31.5 +/- 2.3 ms) and narrower in SkM1 (22.6 +/- 2.8 ms; P=0.03). Premature stimulation induced ventricular tachyarrhythmia/fibrillation >60 seconds in 6 of 8 shams versus 2 of 12 SkM1 (P=0.02). Microelectrode studies of epicardial border zones from SkM1 showed membrane potentials equal to that of shams and V(max) greater than that of shams as membrane potential depolarized (P<0.01). Infarct sizes were similar (sham, 30 +/- 2.8%; SkM1, 30 +/- 2.6%; P=0.86). SkM1 expression in injected epicardium was confirmed immunohistochemically. CONCLUSIONS: SkM1 increases V(max) of depolarized myocardium and reduces the incidence of inducible sustained ventricular tachyarrhythmia/fibrillation in canine infarcts. Gene therapy to normalize activation by increasing V(max) at depolarized potentials may be a promising antiarrhythmic strategy.


Asunto(s)
Terapia Genética/métodos , Sistema de Conducción Cardíaco/fisiología , Modelos Cardiovasculares , Canales de Sodio/genética , Taquicardia Ventricular/fisiopatología , Taquicardia Ventricular/terapia , Potenciales de Acción/fisiología , Adenoviridae/genética , Animales , Línea Celular , Simulación por Computador , Modelos Animales de Enfermedad , Perros , Expresión Génica , Proteínas Fluorescentes Verdes/genética , Humanos , Técnicas In Vitro , Riñón/citología , Masculino , Músculo Esquelético/fisiología , Contracción Miocárdica/fisiología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Infarto del Miocardio/terapia , Canal de Sodio Activado por Voltaje NAV1.5 , Penicilina G/metabolismo , Pericardio/fisiología , Canales de Sodio/metabolismo , Canales de Sodio/fisiología , Taquicardia Ventricular/patología
12.
J Pharmacol Exp Ther ; 332(2): 413-20, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19915070

RESUMEN

Our recent studies demonstrated that propargylamine derivatives such as rasagiline (Azilect, Food and Drug Administration-approved anti-Parkinson drug) and its S-isomer TVP1022 protect cardiac and neuronal cell cultures against apoptotic-inducing stimuli. Studies on structure-activity relationship revealed that their neuroprotective effect is associated with the propargylamine moiety, which protects mitochondrial viability and prevents apoptosis by activating Bcl-2 and protein kinase C-epsilon and by down-regulating the proapoptotic protein Bax. Based on the established cytoprotective and neuroprotective efficacies of propargylamine derivatives, as well as on our recent study showing that TVP1022 attenuates serum starvation-induced and doxorubicin-induced apoptosis in neonatal rat ventricular myocytes (NRVMs), we tested the hypothesis that TVP1022 will also provide protection against doxorubicin-induced NRVM functional derangements. The present study demonstrates that pretreatment of NRVMs with TVP1022 (1 microM, 24 h) prevented doxorubicin (0.5 microM, 24 h)-induced elevation of diastolic [Ca(2+)](i), the slowing of [Ca(2+)](i) relaxation kinetics, and the decrease in the rates of myocyte contraction and relaxation. Furthermore, pretreatment with TVP1022 attenuated the doxorubicin-induced reduction in the protein expression of sarco/endoplasmic reticulum calcium (Ca(2+)) ATPase, Na(+)/Ca(2+) exchanger 1, and total connexin 43. Finally, TVP1022 diminished the inhibitory effect of doxorubicin on gap junctional intercellular coupling (measured by means of Lucifer yellow transfer) and on conduction velocity, the amplitude of the activation phase, and the maximal rate of activation (dv/dt(max)) measured by the Micro-Electrode-Array system. In summary, our results indicate that TVP1022 acts as a novel cardioprotective agent against anthracycline cardiotoxicity, and therefore potentially can be coadmhence, theinistered with doxorubicin in the treatment of malignancies in humans.


Asunto(s)
Cardiotónicos/farmacología , Doxorrubicina/antagonistas & inhibidores , Indanos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Animales , Animales Recién Nacidos , Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Cardiotoxinas/farmacología , Células Cultivadas , Conexina 43/metabolismo , Doxorrubicina/efectos adversos , Uniones Intercelulares/efectos de los fármacos , Contracción Miocárdica/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
13.
J Cardiovasc Electrophysiol ; 21(11): 1276-83, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20487124

RESUMEN

UNLABELLED: Effect of Stretch on Conduction and Cx43. INTRODUCTION: In disease states such as heart failure, myocardial infarction, and hypertrophy, changes in the expression and location of Connexin43 (Cx43) occur (Cx43 remodeling), and may predispose to arrhythmias. Stretch may be an important stimulus to Cx43 remodeling; however, it has only been investigated in neonatal cell cultures, which have different physiological properties than adult myocytes. We hypothesized that localized stretch in vivo causes Cx43 remodeling, with associated changes in conduction, mediated by the renin-angiotensin system (RAS). METHODS AND RESULTS: In an open-chest canine model, a device was used to stretch part of the right ventricle (RV) by 22% for 6 hours. Activation mapping using a 312-electrode array was performed before and after stretch. Regional stretch did not change longitudinal conduction velocity (post-stretch vs baseline: 51.5 ± 5.2 vs 55.3 ± 8.1 cm/s, P = 0.24, n = 11), but significantly reduced transverse conduction velocity (28.7 ± 2.5 vs 35.4 ± 5.4 cm/s, P < 0.01). It also reduced total Cx43 expression, by Western blotting, compared with nonstretched RV of the same animal (86.1 ± 32.2 vs 100 ± 19.4%, P < 0.02, n = 11). Cx43 labeling redistributed to the lateral cell borders. Stretch caused a small but significant increase in the proportion of the dephosphorylated form of Cx43 (stretch 9.95 ± 1.4% vs control 8.74 ± 1.2%, P < 0.05). Olmesartan, an angiotensin II blocker, prevented the stretch-induced changes in Cx43 levels, localization, and conduction. CONCLUSION: Myocardial stretch in vivo has opposite effects to that in neonatal myocytes in vitro. Stretch in vivo causes conduction changes associated with Cx43 remodeling that are likely caused by local stretch-induced activation of the RAS.


Asunto(s)
Conexina 43/metabolismo , Sistema de Conducción Cardíaco/fisiología , Contracción Miocárdica/fisiología , Conducción Nerviosa/fisiología , Sistema Renina-Angiotensina/fisiología , Función Ventricular Derecha/fisiología , Animales , Perros , Módulo de Elasticidad/fisiología , Regulación de la Expresión Génica/fisiología , Distribución Tisular
14.
J Mol Cell Cardiol ; 44(1): 4-13, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17935733

RESUMEN

Gap junctions provide direct cytoplasmic continuity between cells forming a low resistivity barrier to electrical propagation. As such, aberrant regulation of these low resistive conduits has been blamed for electrical conduction disorders in diseased myocardium. While there is a plethora of evidence that abnormalities in gap junctional communication underlie many forms of ventricular arrhythmias, the role of gap junctions in atrial conduction disorders has been less well studied. The atria are the most heterogeneous cardiac structures in terms of the gap junction proteins, connexins (Cx), which are present. Cx40 is the primary, or most abundant, gap junction protein in atria although Cx43 is also abundantly expressed. Cx45 is also expressed in atria, although at low levels. This heterogeneity in connexins leads to a complexity that makes understanding the role of cell coupling in conduction disorders and arrhythmogenesis difficult. In this review we focus on what is known about atrial connexins and their role in atrial fibrillation but also on the challenges presented in understanding the complex interplay between the individual connexin isoforms.


Asunto(s)
Fibrilación Atrial/metabolismo , Conexinas/metabolismo , Animales , Comunicación Celular , Uniones Comunicantes/química , Uniones Comunicantes/metabolismo , Humanos
15.
Prog Biophys Mol Biol ; 94(1-2): 29-65, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17507078

RESUMEN

Gap junctions (GJ), specialised membrane structures that mediate cell-to-cell communication in almost all animal tissues, are composed of intercellular channel-forming integral membrane proteins termed connexins (Cxs), innexins or pannexins. The activity of these channels is closely regulated, particularly by intramolecular modifications as phosphorylation of proteins, via the formation of multiprotein complexes where pore-forming subunits bind to auxiliary channel subunits and associate with scaffolding proteins that play essential roles in channel localization and activity. Scaffolding proteins link signalling enzymes, substrates, and potential effectors (such as channels) into multiprotein signalling complexes that may be anchored to the cytoskeleton. Protein-protein interactions play essential roles in channel localization and activity and, besides their cell-to-cell channel-forming functions, gap junctional proteins now appear involved in different cellular functions (e.g. transcriptional and cytoskeletal regulation). The present review summarizes the recent progress regarding the proteins capable of interacting with junctional proteins and their functional importance.


Asunto(s)
Comunicación Celular/fisiología , Membrana Celular/química , Membrana Celular/metabolismo , Conexinas/química , Conexinas/metabolismo , Uniones Comunicantes/química , Uniones Comunicantes/metabolismo , Modelos Biológicos
16.
Cardiovasc Res ; 75(4): 758-69, 2007 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-17588552

RESUMEN

OBJECTIVE: Atrial fibrillation is often initiated by bursts of ectopic activity arising in the pulmonary veins. We have previously shown that a 3-h intermittent burst pacing protocol (BPP), mimicking ectopic pulmonary vein foci, shortens action potential duration (APD) locally at the pulmonary vein-atrial interface (PV) while having no effect elsewhere in rabbit atrium. This shortening is Ca(2+) dependent and is prevented by apamin, which blocks small conductance Ca(2+)-activated K(+) channels (SK(Ca)). The present study investigates the ionic and molecular mechanisms whereby two apamin-sensitive SK(Ca) channels, SK2 and SK3, might contribute to the regional APD changes. METHODS: Microelectrode and patch clamp techniques were used to record APDs and apamin-sensitive currents in isolated rabbit left atria and cells dispersed from PV and Bachmann's bundle (BB) regions. SK2 and SK3 mRNA and protein levels were quantified, and immunofluorescence was used to observe channel protein distribution. RESULTS: There was a direct relationship between APD shortening and apamin-sensitive current in burst-paced but not sham-paced PV. Moreover, apamin-sensitive current density increased in PV but not BB after BPP. SK2 mRNA, protein, and current were increased in PV after BPP, while SK2 immunostaining shifted from a perinuclear pattern in sham atria to predominance at sites near or at the PV membrane. CONCLUSIONS: BPP-induced acceleration of repolarization in PV results from SK2 channel trafficking to the membrane, leading to increased apamin-sensitive outward current. This is the first indication of involvement of Ca(2+)-activated K(+) currents in atrial remodeling and provides a possible basis for evolution of an arrhythmogenic substrate.


Asunto(s)
Potenciales de Acción/fisiología , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Venas Pulmonares/fisiopatología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Potenciales de Acción/efectos de los fármacos , Animales , Apamina/farmacología , Transporte Biológico , Membrana Celular/metabolismo , Atrios Cardíacos , Inmunohistoquímica , Masculino , Microelectrodos , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Venas Pulmonares/metabolismo , ARN Mensajero/análisis , Conejos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/análisis , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/genética
17.
Cardiovasc Res ; 72(2): 241-9, 2006 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-16914125

RESUMEN

BACKGROUND: The epicardial border zone (EBZ) of surviving myocytes in the healing, 4- to 5-day-old canine infarct is an arrhythmogenic substrate characterized by both structural and functional remodeling of Cx43. Unknown is whether the remodeling of gap junction conductance is heterogeneous in the EBZ like that of sarcolemmal ion channel remodeling and how remodeling of the gap junction influences conduction and anisotropy. METHODS AND RESULTS: Ventricular tachycardia was initiated by programmed stimulation in healing canine infarcted hearts. Reentrant circuits were mapped and the central common pathway (CCP) and outer pathway (OP) regions localized. Epimyocardium removed from the CCP was disaggregated to generate myocyte pairs for conductance measurements. Cx43 distribution was determined by immunofluorescent confocal microscopy. While transverse coupling (gap junction conductance) was markedly decreased in OP cells, CCP cells with lateralized Cx43 gap junctions showed normal conductance. Longitudinal coupling in both OP and CCP was no different than normal. Consistent with conductance measurements, the anisotropic ratio in the CCP was similar to that of normal tissue. In the OP it was increased. Despite normal longitudinal and transverse conductance and anisotropic ratio, longitudinal and transverse conduction velocities were decreased in the CCP with respect to normal epicardium, possibly as a result of the remodeling of sarcolemmal ion channels in this region. CONCLUSIONS: Gap junction conductance and distribution is heterogeneous in different regions of reentrant circuits. Lateralization of Cx43 gap junctions in CCP of reentrant circuits is associated with normal transverse conductance between cell pairs. In contrast, absence of lateralization in OP is associated with reduced transverse conductance. Despite normal anisotropic ratio, conduction velocity in CCP region remains slower than normal. This suggests that the effects of Cx43 remodeling in the infarcted heart should be interpreted in conjunction with other types of remodeling occurring in the EBZ (i.e. sarcolemmal ion channels).


Asunto(s)
Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Infarto del Miocardio/metabolismo , Pericardio/metabolismo , Animales , Anisotropía , Estimulación Cardíaca Artificial , Conexina 43/análisis , Perros , Electrocardiografía , Microscopía Confocal , Miocitos Cardíacos/metabolismo , Taquicardia por Reentrada en el Nodo Atrioventricular/metabolismo
19.
Circ Res ; 95(4): e22-8, 2004 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-15284189

RESUMEN

Connexin43 (Cx43) channels reside in at least 3 states: closed, open, or residual. It is hypothesized that the residual state results from the interaction of an intracellular "gating element" with structures at the vestibule of the pore. Recently, we showed in vitro that there is an intramolecular interaction of the carboxyl-terminal domain (referred to as "CT") with a region in the cytoplasmic loop of Cx43 (amino acids 119 to 144; referred to as "L2"). Here, we assessed whether the L2 region was able to interact with the gating particle in a functional channel. Cx43 channels were recorded in the presence of a peptide corresponding to the L2 region, delivered via the patch pipette. This manipulation did not modify unitary conductance, but decreased the frequency of transitions into the residual state, prolonged open time, and altered the voltage dependence of the channel in a manner analogous to that observed after truncation of the CT domain. The latter correlated with the ability of the peptide to bind to the CT domain, as determined by mirror resonance spectroscopy. Overall, we propose that the L2 acts as a "receptor" that interacts with a flexible intracellular gating element during channel gating. The full text of this article is available online at http://circres.ahajournals.org.


Asunto(s)
Conexina 43/química , Uniones Comunicantes/fisiología , Activación del Canal Iónico/fisiología , Animales , Línea Celular Tumoral/patología , Conexina 43/genética , Uniones Comunicantes/efectos de los fármacos , Genes Reporteros , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Activación del Canal Iónico/efectos de los fármacos , Ratones , Mutación , Neuroblastoma/patología , Fragmentos de Péptidos/síntesis química , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/farmacología , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Relación Estructura-Actividad , Transfección
20.
Circ Res ; 94(2): 215-22, 2004 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-14699011

RESUMEN

Ischemia-induced acidification of astrocytes or cardiac myocytes reduces intercellular communication by closing gap junction channels and subsequently internalizing gap junction proteins. To determine whether such coupling changes might be attributable to altered interactions between connexin43 (Cx43) and other proteins, we applied the nigericin/high K+ method to vary intracellular pH (pHi) in cultured cortical astrocytes. Intracellular acidification was accompanied by internalization of Cx43 with retention of Cx43 scaffolding protein Zonula Occludens-1 (ZO-1) at cell surfaces, suggesting that ZO-1 and Cx43 dissociate at low pHi. Coimmunoprecipitation studies revealed decreased binding of ZO-1 and increased binding of c-Src to Cx43 at low pHi. Resonant mirror spectroscopy was used to quantify binding of the SH3 domain of c-Src and the PDZ domains of ZO-1 to the carboxyl terminal domain of Cx43 (Cx43CT). Data indicate that the c-Src/Cx43CT interaction is highly pH dependent whereas the ZO-1/Cx43CT interaction is not. Moreover, binding of c-Src to Cx43CT prevented and reversed ZO-1/Cx43CT binding. We hypothesize that increased affinity of c-Src for Cx43 at low pHi aids in separation of Cx43 from ZO-1 and that this may facilitate internalization of Cx43. These data suggest that protracted acidification may remodel protein-protein interactions involving Cx43 and thus provide an important protective mechanism to limit lesion spread after ischemic injury.


Asunto(s)
Astrocitos/metabolismo , Conexina 43/metabolismo , Uniones Comunicantes/metabolismo , Concentración de Iones de Hidrógeno , Proteínas de la Membrana/metabolismo , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/ultraestructura , Células Cultivadas/efectos de los fármacos , Células Cultivadas/metabolismo , Células Cultivadas/ultraestructura , Conexina 43/química , Uniones Comunicantes/efectos de los fármacos , Líquido Intracelular/química , Isquemia/metabolismo , Cinética , Sustancias Macromoleculares , Proteínas de la Membrana/química , Ratones , Ratones Endogámicos C57BL , Nigericina/farmacología , Fosfoproteínas/química , Potasio/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas pp60(c-src)/química , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteína de la Zonula Occludens-1 , Dominios Homologos src
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA