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








Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 21: 369-381, 2021 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-33898634

RESUMO

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.

2.
Cell Mol Life Sci ; 72(15): 2779-92, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26091747

RESUMO

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.


Assuntos
Sistema Cardiovascular/metabolismo , Conexinas/metabolismo , Animais , Fenômenos Fisiológicos Cardiovasculares , Junções Comunicantes/metabolismo , Junções Comunicantes/fisiologia , Humanos , Canais Iônicos/metabolismo
3.
J Biol Chem ; 288(43): 31139-53, 2013 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-24022490

RESUMO

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.


Assuntos
Trifosfato de Adenosina/metabolismo , Eritrócitos/metabolismo , Capeamento Imunológico , Receptores de Complemento 3b/metabolismo , Trifosfato de Adenosina/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Eritrócitos/citologia , Eritrócitos/imunologia , Feminino , Humanos , Masculino , Lipídeos de Membrana/imunologia , Lipídeos de Membrana/metabolismo , Fagocitose/imunologia , Receptores de Complemento 3b/imunologia
4.
Circ Arrhythm Electrophysiol ; 6(3): 623-31, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23559673

RESUMO

BACKGROUND: Previously, we showed that a mouse model (ACE8/8) of cardiac renin-angiotensin system activation has a high rate of spontaneous ventricular tachycardia and sudden cardiac death secondary to a reduction in connexin43 level. Angiotensin-II activation increases reactive oxygen species (ROS) production, and ACE8/8 mice show increased cardiac ROS. We sought to determine the source of ROS and whether ROS played a role in the arrhythmogenesis. METHODS AND RESULTS: Wild-type and ACE8/8 mice with and without 2 weeks of treatment with L-NIO (NO synthase inhibitor), sepiapterin (precursor of tetrahydrobiopterin), MitoTEMPO (mitochondria-targeted antioxidant), TEMPOL (a general antioxidant), apocynin (nicotinamide adenine dinucleotide phosphate oxidase inhibitor), allopurinol (xanthine oxidase inhibitor), and ACE8/8 crossed with P67 dominant negative mice to inhibit the nicotinamide adenine dinucleotide phosphate oxidase were studied. Western blotting, detection of mitochondrial ROS by MitoSOX Red, electron microscopy, immunohistochemistry, fluorescent dye diffusion technique for functional assessment of connexin43, telemetry monitoring, and in vivo electrophysiology studies were performed. Treatment with MitoTEMPO reduced sudden cardiac death in ACE8/8 mice (from 74% to 18%; P<0.005), decreased spontaneous ventricular premature beats, decreased ventricular tachycardia inducibility (from 90% to 17%; P<0.05), diminished elevated mitochondrial ROS to the control level, prevented structural damage to mitochondria, resulted in 2.6-fold increase in connexin43 level at the gap junctions, and corrected gap junction conduction. None of the other antioxidant therapies prevented ventricular tachycardia and sudden cardiac death in ACE8/8 mice. CONCLUSIONS: Mitochondrial oxidative stress plays a central role in angiotensin II-induced gap junction remodeling and arrhythmia. Mitochondria-targeted antioxidants may be effective antiarrhythmic drugs in cases of renin-angiotensin system activation.


Assuntos
Antioxidantes/farmacologia , Conexina 43/metabolismo , Morte Súbita Cardíaca/etiologia , Mitocôndrias/metabolismo , Estresse Oxidativo/fisiologia , Taquicardia Ventricular/tratamento farmacológico , Acetofenonas/farmacologia , Animais , Conexina 43/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos , NADPH Oxidases/farmacologia , Distribuição Aleatória , Espécies Reativas de Oxigênio/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Fatores de Risco , Sensibilidade e Especificidade , Marcadores de Spin , Taquicardia Ventricular/fisiopatologia
5.
Am J Physiol Heart Circ Physiol ; 303(10): H1208-18, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22982782

RESUMO

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.


Assuntos
Trifosfato de Adenosina/metabolismo , Conexinas/metabolismo , Fibroblastos/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Comunicação Parácrina , Animais , Membrana Celular/metabolismo , Técnicas de Cocultura , Conexinas/genética , Modelos Animais de Doenças , Cães , Fibroblastos/patologia , Fibrose , Glicosilação , Células Madin Darby de Rim Canino , Camundongos , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Miócitos Cardíacos/patologia , Miofibroblastos/metabolismo , Miofibroblastos/patologia , Proteínas do Tecido Nervoso/genética , Fenótipo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Transporte Proteico , Sarcolema/metabolismo , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
6.
Front Physiol ; 3: 272, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22934026

RESUMO

BACKGROUND: The proinflammatory cytokine Interleukin-1ß (IL-1ß), which increases in the heart post myocardial infarction (MI), has been shown to cause loss of Connexin43 (Cx43) function, an event known to underlie formation of the arrhythmogenic substrate. Omega 3 Fatty acids exhibit antiarrhythmic properties and impact IL-1ß signaling. We hypothesize that Omega-3 fatty acids prevent arrhythmias in part, by inhibiting IL-1ß signaling thus maintaining functional Cx43 channels. METHODS: Rat neonatal myocytes or Madin-Darby Canine Kidney Epithelial (MDCK) cells grown in media in the absence (Ctr) or presence of 30 µM docosahexaenoic acid (DHA, an Omega-3 Fatty acid) were treated with 0.1 µM activated IL-1ß. We determined Cx43 channel function using a dye spread assay. Western blot and immunostaining were used to examine Cx43 levels/localization and downstream effectors of IL-1ß. In addition we used a murine model of MI for 24 h to determine the impact of an Omega-3 fatty acid enriched diet on Cx43 levels/localization post MI. RESULTS: IL-1ß significantly inhibited Cx43 function in Ctr cells (200.9 ± 17.7 µm [Ctr] vs. 112.8 ± 14.9 µm [0.1 uM IL-1ß], p<0.05). However, DHA-treated cells remained highly coupled in the presence of IL-1ß [167.9 ± 21.9 µm [DHA] vs. 164.4 ± 22.3 µm [DHA + 0.1 uM IL-1ß], p<0.05, n = 4]. Additionally, western blot showed that IL-1ß treatment caused a 38.5% downregulation of Cx43 [1.00 au [Ctr] vs. 0.615 au (0.1 µM IL-1ß) which was completely abolished in DHA-treated cells (0.935 au [DHA] vs. 1.02 au [DHA + 0.1 µM IL-1ß), p < 0.05, n = 3]. Examination of the downstream modulator of IL-1ß, NFκß showed that while hypoxia caused translocation of NFκß to the nucleus, this was inhibited by DHA. Additionally we found that a diet enriched in Omega-3 Fatty acids inhibited lateralization of Cx43 in the post-MI murine heart as well as limited activation of fibroblasts which would lead to decreased fibrosis overall. CONCLUSIONS: Omega 3 Fatty acid treatment inhibited IL-1ß-stimulated loss of Cx43 protein, and more importantly, inhibited loss of Cx43 function by inhibiting translocation of NFκß. In the intact heart a diet enriched in Omega 3 Fatty Acids limited loss of Cx43 at the intercalated disk in the heart following MI. These data suggest that one of cardio-protective mechanisms by which Omega 3 Fatty acids work includes prevention of the pro-arrhythmic loss of Cx43 post MI and the attenuation of cardiac fibrosis after injury.

7.
Arterioscler Thromb Vasc Biol ; 32(8): 1951-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22652601

RESUMO

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.


Assuntos
Comunicação Celular , Conexinas/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Células Endoteliais/fisiologia , Proteínas Musculares/metabolismo , Proteínas Musculares/fisiologia , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Animais , Agregação Celular , Células Cultivadas , Conexina 43/genética , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Proteínas Musculares/genética , Neovascularização Fisiológica , Fatores de Transcrição de Domínio TEA , Fatores de Transcrição/genética
8.
Cardiovasc Res ; 94(3): 450-9, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22374989

RESUMO

AIMS: Reentry accounts for most life-threatening arrhythmias, complicating myocardial infarction, and therapies that consistently prevent reentry from occurring are lacking. In this study, we compare antiarrhythmic effects of gene transfer of green fluorescent protein (GFP; sham), the skeletal muscle sodium channel (SkM1), the liver-specific connexin (Cx32), and SkM1/Cx32 in the subacute canine infarct. METHODS AND RESULTS: Immediately after ligation of the left anterior descending artery, viral constructs were implanted in the epicardial border zone (EBZ). Five to 7 days later, efficient restoration of impulse propagation (narrow QRS and local electrogram duration) occurred in SkM1, Cx32, and SkM1/Cx32 groups (P< 0.05 vs. GFP). Programmed electrical stimulation from the EBZ induced sustained ventricular tachycardia (VT)/ventricular fibrillation (VF) in 15/22 GFP dogs vs. 2/12 SkM1, 6/14 Cx32, and 8/10 SkM1/Cx32 (P< 0.05 SkM1 vs. GFP). GFP, SkM1, and SkM1/Cx32 had predominantly polymorphic VT/VF, whereas in Cx32 dogs, monomorphic VT predominated (P< 0.05 for Cx32 vs. GFP). Tetrazolium red staining showed significantly larger infarcts in Cx32- vs. GFP-treated animals (P< 0.05). CONCLUSION: Whereas SkM1 gene transfer reduces the incidence of inducible VT/VF, Cx32 therapy to improve gap junctional conductance results in larger infarct size, a different VT morphology, and no antiarrhythmic efficacy.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Conexinas/metabolismo , Junções Comunicantes/efeitos dos fármacos , Proteínas Musculares/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Canais de Sódio/metabolismo , Fibrilação Ventricular/tratamento farmacológico , Animais , Antiarrítmicos/uso terapêutico , Conexinas/genética , Cães , Estimulação Elétrica , Eletrocardiografia , Masculino , Camundongos , Proteínas Musculares/genética , Ratos , Canais de Sódio/genética , Fibrilação Ventricular/fisiopatologia , Proteína beta-1 de Junções Comunicantes
9.
Circulation ; 125(3): 474-81, 2012 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-22179534

RESUMO

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.


Assuntos
Conexina 43/genética , Junções Comunicantes/patologia , Junções Comunicantes/fisiologia , Mutação de Sentido Incorreto , Morte Súbita do Lactente/genética , Morte Súbita do Lactente/patologia , Adulto , Animais , Caderinas/metabolismo , Estudos de Coortes , Conexina 43/metabolismo , Desmoplaquinas/metabolismo , Feminino , Predisposição Genética para Doença/genética , Humanos , Lactente , Masculino , Miócitos Cardíacos/patologia , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Transporte Proteico/genética , Ratos
11.
Am J Physiol Heart Circ Physiol ; 302(3): H790-800, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22101526

RESUMO

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.


Assuntos
Comunicação Celular/fisiologia , Conexina 43/metabolismo , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Cicatrização/fisiologia , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/patologia , Linhagem Celular , Simulação por Computador , Cães , Corantes Fluorescentes/farmacocinética , Junções Comunicantes/fisiologia , Interleucina-1beta/metabolismo , Isoquinolinas/farmacocinética , Rim/citologia , Modelos Cardiovasculares , Infarto do Miocárdio/patologia , Miócitos Cardíacos/citologia , Miofibroblastos/citologia , Comunicação Parácrina/fisiologia , Ratos
12.
J Am Coll Cardiol ; 58(22): 2332-9, 2011 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-22093512

RESUMO

OBJECTIVES: The aim of this study was to test whether c-Src tyrosine kinase mediates connexin-43 (Cx43) reduction and sudden cardiac death in a transgenic mouse model of cardiac-restricted overexpression of angiotensin-converting enzyme (ACE8/8 mice). BACKGROUND: Renin-angiotensin system activation is associated with an increased risk for arrhythmia and sudden cardiac death, but the mechanism is not well understood. The up-regulation of c-Src by angiotensin II may result in the reduction of Cx43, which impairs gap junction function and provides a substrate for arrhythmia. METHODS: Wild-type and ACE8/8 mice with and without treatment with the c-Src inhibitor 1-(1,1-dimethylethyl)-1-(4-methylphenyl)-1H-pyrazolo[3,4-d]pyrimidin-4-amine (PP1) were studied. Telemetry monitoring, in vivo electrophysiologic studies, Western blot analyses for total and phosphorylated c-Src and Cx43, immunohistochemistry staining for Cx43, and functional assessment of Cx43 with fluorescent dye diffusion were performed. RESULTS: The majority of the arrhythmic deaths resulted from ventricular tachycardia degenerating to ventricular fibrillation (83%). Levels of total and phosphorylated c-Src were increased and Cx43 reduced in ACE8/8 mice. PP1 reduced total and phosphorylated c-Src levels, increased Cx43 level by 2.1-fold (p < 0.005), increased Cx43 at the gap junctions (immunostaining), improved gap junctional communication (dye spread), and reduced ventricular tachycardia inducibility and sudden cardiac death. The survival rate increased from 11% to 86% with 4 weeks of PP1 treatment (p < 0.005). Treatment with an inactive analog did not change survival or Cx43 levels. CONCLUSIONS: Renin-angiotensin system activation is associated with c-Src up-regulation, Cx43 loss, reduced myocyte coupling, and arrhythmic sudden death, which can be prevented by c-Src inhibition. This suggests that an increase in c-Src activity may help mediate renin-angiotensin system-induced arrhythmias and that c-Src inhibitors might exert antiarrhythmic activity.


Assuntos
Angiotensina II/metabolismo , Conexina 43/metabolismo , Morte Súbita Cardíaca/prevenção & controle , Junções Comunicantes/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Animais , Western Blotting , Proteína Tirosina Quinase CSK , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fosforilação , Pirazóis/farmacologia , Pirimidinas/farmacologia , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/prevenção & controle , Telemetria , Fibrilação Ventricular/metabolismo , Fibrilação Ventricular/prevenção & controle , Quinases da Família src
13.
Channels (Austin) ; 5(3): 193-7, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21532340

RESUMO

Pannexins, a class of membrane channels, bear significant sequence homology with the invertebrate gap junction proteins, innexins and more distant similarities in their membrane topologies and pharmacological sensitivities with the gap junction proteins, connexins. However, the functional role for the pannexin oligomers, or pannexons, is different from connexin oligomers, the connexons. Many pannexin publications have used the term "hemichannels" to describe pannexin oligomers while others use the term "channels" instead. This has led to confusion within the literature about the function of pannexins that promotes the idea that pannexons serve as gap junction hemichannels and thus have an assembly and functional state as gap junctional intercellular channels. Here we present the case that unlike the connexin gap junction intercellular channels, so far, pannexin oligomers have repeatedly been shown to be channels that are functional in single membranes, but not as intercellular channel in appositional membranes. Hence, they should be referred to as channels and not hemichannels. Thus, we advocate that in the absence of firm evidence that pannexins form gap junctions, the use of the term "hemichannel" be discontinued within the pannexin literature.


Assuntos
Conexinas/metabolismo , Junções Comunicantes/metabolismo , Canais Iônicos/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Multimerização Proteica/fisiologia , Animais , Conexinas/genética , Junções Comunicantes/genética , Humanos , Canais Iônicos/genética , Proteínas do Tecido Nervoso/genética , Publicações Periódicas como Assunto , Terminologia como Assunto
14.
J Mol Med (Berl) ; 89(7): 677-87, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21553032

RESUMO

Renin-angiotensin system (RAS) activation is associated with arrhythmias. We investigated the effects of RAS inhibition in cardiac-specific angiotensin-converting enzyme (ACE) overexpression (ACE 8/8) mice, which exhibit proclivity to ventricular tachycardia (VT) and sudden death because of reduced connexin43 (Cx43). ACE 8/8 mice were treated with an ACE inhibitor (captopril) or an angiotensin receptor type-1 blocker (losartan). Subsequently, electrophysiological studies were performed, and the hearts were extracted for Cx43 quantification using immunoblotting, immunohistochemistry, fluorescent dye spread method, and sodium current quantification using whole cell patch clamping. VT was induced in 12.5% of captopril-treated ACE 8/8 and in 28.6% of losartan-treated mice compared to 87.5% of untreated mice (P < 0.01). Losartan and captopril treatment increased total Cx43 2.4-fold (P = 0.01) and the Cx43 phosphorylation ratio 2.3-fold (P = 0.005). Treatment was associated with a recovery of gap junctional conductance. Survival in treated mice improved to 0.78 at 10 weeks (95% confidence interval 0.64 to 0.92), compared to the expected survival of less than 0.50. In a model of RAS activation, arrhythmic risk was correlated with reduced Cx43 amount and phosphorylation. RAS inhibition resulted in increased total and phosphorylated Cx43, decreased VT inducibility, and improved survival.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Conexina 43/metabolismo , Sistema Renina-Angiotensina , Risco , Taquicardia Ventricular/fisiopatologia , Angiotensina II/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Eletrocardiografia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/metabolismo , Fosforilação/efeitos dos fármacos , Taquicardia Ventricular/metabolismo
15.
J Clin Invest ; 121(5): 1894-904, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21505261

RESUMO

The epicardium makes essential cellular and paracrine contributions to the growth of the fetal myocardium and the formation of the coronary vasculature. However, whether the epicardium has similar roles postnatally in the normal and injured heart remains enigmatic. Here, we have investigated this question using genetic fate-mapping approaches in mice. In uninjured postnatal heart, epicardial cells were quiescent. Myocardial infarction increased epicardial cell proliferation and stimulated formation of epicardium-derived cells (EPDCs), which remained in a thickened layer on the surface of the heart. EPDCs did not adopt cardiomyocyte or coronary EC fates, but rather differentiated into mesenchymal cells expressing fibroblast and smooth muscle cell markers. In vitro and in vivo assays demonstrated that EPDCs secreted paracrine factors that strongly promoted angiogenesis. In a myocardial infarction model, EPDC-conditioned medium reduced infarct size and improved heart function. Our findings indicate that epicardium modulates the cardiac injury response by conditioning the subepicardial environment, potentially offering a new therapeutic strategy for cardiac protection.


Assuntos
Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Pericárdio/metabolismo , Animais , Fibroblastos/metabolismo , Coração/fisiologia , Traumatismos Cardíacos/metabolismo , Homeostase , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Camundongos , Modelos Biológicos , Modelos Genéticos , Infarto do Miocárdio/patologia , Miocárdio/metabolismo , Neovascularização Patológica , Resultado do Tratamento
16.
Circ Arrhythm Electrophysiol ; 4(3): 344-51, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21493965

RESUMO

BACKGROUND: The border zone of healing myocardial infarcts is an arrhythmogenic substrate, partly the result of structural and functional remodeling of the ventricular gap junction protein, Connexin43 (Cx43). Cx43 in arrhythmogenic substrates is a potential target for antiarrhythmic therapy. METHODS AND RESULTS: We characterized Cx43 remodeling in the epicardial border zone (EBZ) of healing canine infarcts 5 days after coronary occlusion and examined whether the gap junction-specific agent rotigaptide could reverse it. Cx43 remodeling in the EBZ was characterized by a decrease in Cx43 protein, lateralization, and increased Cx43 phosphorylation at serine (S) 368. Rotigaptide partially reversed the loss of Cx43 but did not affect the increase in S368 phosphorylation, nor did it reverse Cx43 lateralization. Rotigaptide did not prevent conduction slowing in the EBZ, nor did it decrease the induction of sustained ventricular tachycardia by programmed stimulation, although it did decrease the EBZ effective refractory period. CONCLUSIONS: We conclude that partial reversal of Cx43 remodeling in healing infarct border zone may not be sufficient to restore normal conduction or prevent arrhythmias.


Assuntos
Fenômenos Eletrofisiológicos/fisiologia , Junções Comunicantes/fisiologia , Infarto do Miocárdio/complicações , Oligopeptídeos/farmacologia , Pericárdio/metabolismo , Recuperação de Função Fisiológica/fisiologia , Taquicardia Ventricular/metabolismo , Animais , Modelos Animais de Doenças , Cães , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Junções Comunicantes/efeitos dos fármacos , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Pericárdio/fisiopatologia , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/etiologia
17.
Front Physiol ; 2: 1, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21423411

RESUMO

Increasing connexin43 (Cx43) gap junctional conductance as a means to improve cardiac conduction has been proposed as a novel antiarrhythmic modality. Yet, transmission of molecules via gap junctions may be associated with increased infarct size. To determine whether maintaining open gap junction channels impacts on infarct size and induction of ventricular tachycardia (VT) following coronary occlusion, we expressed the pH- and voltage-independent connexin isoform connexin32 (Cx32) in ventricle and confirmed Cx32 expression. Wild-type (WT) mice injected with adenovirus-Cx32 (Cx32inj) were examined following coronary occlusion to determine infarct size and inducibility of VT. There was an increased infarct size in Cx32inj hearts as compared to WT (WT 22.9 ± 4%; Cx32inj 44.3 ± 5%; p < 0.05). Programmed electrical stimulation showed no difference in VT inducibility in WT and Cx32inj mice (VT was reproducibly inducible in 55% of shams and 50% of Cx32inj mice (p > 0.05). Following coronary occlusion, improving cell-cell communication increased infarct size, and conferred no antiarrhythmic benefit.

18.
J Cardiovasc Pharmacol ; 57(4): 376-9, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21297493

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Miocárdio/citologia , Miofibroblastos/metabolismo , Transdiferenciação Celular , Citocinas/metabolismo , Matriz Extracelular/metabolismo , Humanos , Inflamação/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo
19.
J Cardiovasc Pharmacol ; 57(4): 373-5, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21346592

RESUMO

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.


Assuntos
Fibroblastos/metabolismo , Miócitos Cardíacos/metabolismo , Miofibroblastos/metabolismo , Fibrose , Humanos , Miocárdio/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA