Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Transl Med ; 5(207): 207ra141, 2013 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-24132636

RESUMO

Percutaneous coronary intervention is first-line therapy for acute coronary syndromes (ACS) but can promote cardiomyocyte death and cardiac dysfunction via reperfusion injury, a phenomenon driven in large part by oxidative stress. Therapies to limit this progression have proven elusive, with no major classes of new agents since the development of anti-platelets/anti-thrombotics. We report that cardiac troponin I-interacting kinase (TNNI3K), a cardiomyocyte-specific kinase, promotes ischemia/reperfusion injury, oxidative stress, and myocyte death. TNNI3K-mediated injury occurs through increased mitochondrial superoxide production and impaired mitochondrial function and is largely dependent on p38 mitogen-activated protein kinase (MAPK) activation. We developed a series of small-molecule TNNI3K inhibitors that reduce mitochondrial-derived superoxide generation, p38 activation, and infarct size when delivered at reperfusion to mimic clinical intervention. TNNI3K inhibition also preserves cardiac function and limits chronic adverse remodeling. Our findings demonstrate that TNNI3K modulates reperfusion injury in the ischemic heart and is a tractable therapeutic target for ACS. Pharmacologic TNNI3K inhibition would be cardiac-selective, preventing potential adverse effects of systemic kinase inhibition.


Assuntos
MAP Quinase Quinase Quinases/antagonistas & inibidores , Isquemia Miocárdica/enzimologia , Isquemia Miocárdica/fisiopatologia , Estresse Oxidativo , Proteínas Quinases/metabolismo , Remodelação Ventricular , Síndrome Coronariana Aguda/complicações , Síndrome Coronariana Aguda/enzimologia , Síndrome Coronariana Aguda/patologia , Síndrome Coronariana Aguda/fisiopatologia , Animais , Morte Celular/efeitos dos fármacos , Modelos Animais de Doenças , Metabolismo Energético/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Deleção de Genes , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/enzimologia , Insuficiência Cardíaca/fisiopatologia , Humanos , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Isquemia Miocárdica/complicações , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Estresse Oxidativo/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Superóxidos/metabolismo , Regulação para Cima/efeitos dos fármacos , Disfunção Ventricular Esquerda/complicações , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/patologia , Disfunção Ventricular Esquerda/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
2.
Front Pharmacol ; 3: 128, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22783192

RESUMO

Soluble guanylate cyclase (sGC), the primary mediator of nitric oxide (NO) bioactivity, exists as reduced (NO-sensitive) and oxidized (NO-insensitive) forms. We tested the hypothesis that the cardiovascular protective effects of NO-insensitive sGC activation would be potentiated under conditions of oxidative stress compared to those of NO-sensitive sGC stimulation. The cardiovascular effects of the NO-insensitive sGC activator GSK2181236A [a low, non-depressor dose, and a high dose which lowered mean arterial pressure (MAP) by 5-10 mmHg] and those of equi-efficacious doses of the NO-sensitive sGC stimulator BAY 60-4552 were assessed in (1) Sprague Dawley rats during coronary artery ischemia/reperfusion (I/R) and (2) spontaneously hypertensive stroke prone rats (SHR-SP) on a high salt/fat diet (HSFD). In I/R, neither compound reduced infarct size 24 h after reperfusion. In SHR-SP, HSFD increased MAP, urine output, microalbuminuria, and mortality, caused left ventricular hypertrophy with preserved ejection fraction, and impaired endothelium-dependent vasorelaxation. The low dose of BAY 60-4552, but not that of GSK2181236A, decreased urine output, and improved survival. Conversely, the low dose of GSK2181236A, but not that of BAY 60-4552, attenuated the development of cardiac hypertrophy. The high doses of both compounds similarly attenuated cardiac hypertrophy and improved survival. In addition to these effects, the high dose of BAY 60-4552 reduced urine output and microalbuminuria and attenuated the increase in MAP to a greater extent than did GSK2181236A. Neither compound improved endothelium-dependent vasorelaxation. In SHR-SP isolated aorta, the vasodilatory responses to the NO-dependent compounds carbachol and sodium nitroprusside were attenuated by HSFD. In contrast, the vasodilatory responses to both GSK2181236A and BAY 60-4552 were unaltered by HSFD, indicating that reduced NO-bioavailability and not changes in the oxidative state of sGC is responsible for the vascular dysfunction. In summary, GSK2181236A and BAY 60-4552 provide partial benefit against hypertension-induced end-organ damage. The differential beneficial effects observed between these compounds could reflect tissue-specific changes in the oxidative state of sGC and might help direct the clinical development of these novel classes of therapeutic agents.

3.
Heart Fail Rev ; 15(5): 447-56, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20225094

RESUMO

Despite a wide range of therapeutic interventions, the prognosis for most patients with heart failure remains poor. The identification of stem cells with the ability to generate cardiomyocytes and vascular cells and promote local repair and survival pathways has highlighted the ability of the heart to undergo regeneration and potentially provides a new therapeutic strategy for treatment of the failing heart. In recent years, however, clinical trials aimed at exploiting the beneficial effects of stem and progenitor cells to treat patients with cardiovascular disease have resulted in mild improvements at best, suggesting that these cells and/or the conditions in which they find themselves are not conducive to cardiac repair. Heart failure is most prevalent among older individuals, and a growing body of evidence suggests that with increasing age, cardiac stem and progenitor cells undergo senescent changes that impair their regenerative capacities. Moreover, environmental alterations over time appear to impact the capacity of these cells to improve cardiac function. Understanding these senescent changes may lead to the development of new and improved approaches to exploit the potential of stem cells to repair the aging heart. In this review, age-associated alterations in cardiac stem cell function are discussed, as well as strategies that are being investigated to promote cardiac regeneration in the patient with heart failure.


Assuntos
Envelhecimento/patologia , Insuficiência Cardíaca/terapia , Miócitos Cardíacos/transplante , Transplante de Células-Tronco , Células-Tronco , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Transplante de Medula Óssea , Insuficiência Cardíaca/fisiopatologia , Humanos
4.
Aging Cell ; 7(4): 599-604, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18489728

RESUMO

Significant advances in the past decade have revealed that a large number of highly plastic stem cells are maintained in humans through adulthood and are present even in older adults. These findings are notable in light of the reduced capacity for repair and regeneration in older tissues. The apparent dichotomy can be reconciled through an appreciation of the age-associated changes in the microenvironmental pathways that govern adult stem cell plasticity and differentiation patterns. Specifically, the recent identification of the age-related loss of the local platelet-derived growth factor signals that promote the induction of cardiac myocytes from Oct-3/4+ bone marrow stem cells, rather than impairment in the stem cells themselves, provides a template for understanding and targeting the environmental pathways underlying the regenerative capacity of older tissues and organs. It is projected that this paradigm extends to the overall regulation of adult stem cell biology, shifting the balance from tissue generation during development and maturation to the prevention of untoward stem cell differentiation with aging.


Assuntos
Envelhecimento/fisiologia , Meio Ambiente , Células-Tronco/citologia , Células-Tronco Adultas/citologia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular , Humanos
5.
J Mol Cell Cardiol ; 45(4): 582-92, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18396293

RESUMO

The discovery of extracardiac progenitor cells and resident cardiac stem cells in recent years has led to a great deal of interest in the development of therapeutic strategies that target these endogenous cell sources for promotion of cardiovascular repair mechanisms in the diseased heart. Cardiovascular risk increases with age and among many factors, the age-associated decline in cardiac and vascular regenerative capacity may contribute to the progressive deterioration of cardiovascular health. Thus, understanding the mechanisms which underlie the dysregulation of cardiac stem and progenitor cells may lead to the identification of novel targets and approaches to reverse this decline. In this review, we outline the current knowledge about cardiac stem and progenitor cells, their contribution to cardiovascular regenerative processes and factors that may affect their decreased function in aging individuals. Moreover, we describe the therapeutic strategies that are currently being tested in clinical trials as well as potential new avenues of investigation for the future.


Assuntos
Envelhecimento , Doenças Cardiovasculares/terapia , Miocárdio , Transplante de Células-Tronco/métodos , Células-Tronco , Fatores Etários , Animais , Humanos , Fatores de Risco
6.
Expert Opin Ther Targets ; 11(11): 1385-99, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18028005

RESUMO

Vascular dysfunction underlies the pathophysiology of a wide range of diseases, including atherosclerosis, diabetes and arthritis. Angiogenic function is progressively impaired with increasing age and, therefore, has been linked to the increased risk of many of these diseases among older people. Elucidating the cellular and molecular angiogenic pathways that become dysregulated with age will lead to the identification of novel targets for the restoration of vascular repair mechanisms in the older population. This review examines the regulation of postnatal angiogenesis in vascular disease, the changes observed in ageing and highlights potential therapeutic targets, including endothelial progenitor cell-based strategies for the promotion of angiogenic pathways that are impaired with age.


Assuntos
Envelhecimento/fisiologia , Inibidores da Angiogênese/farmacologia , Neovascularização Patológica/tratamento farmacológico , Neovascularização Fisiológica/efeitos dos fármacos , Idoso , Sistemas de Liberação de Medicamentos , Endotélio Vascular/metabolismo , Humanos , Neovascularização Patológica/fisiopatologia , Fatores de Risco , Regulação para Cima , Doenças Vasculares/tratamento farmacológico , Doenças Vasculares/fisiopatologia
7.
Circ Res ; 100(8): 1116-27, 2007 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-17463327

RESUMO

It is well established that cardiovascular repair mechanisms become progressively impaired with age and that advanced age is itself a significant risk factor for cardiovascular disease. Although therapeutic developments have improved the prognosis for those with cardiovascular disease, mortality rates have nevertheless remained virtually unchanged in the last twenty years. Clearly, there is a need for alternative strategies for the treatment of cardiovascular disease. In recent years, the idea that the heart is capable of regeneration has raised the possibility that cell-based therapies may provide such an alternative to conventional treatments. Cells that have the potential to generate cardiomyocytes and vascular cells have been identified in both the adult heart and peripheral tissues, and in vivo experiments suggest that these cardiovascular stem cells and cardiovascular progenitor cells, including endothelial progenitor cells, are capable of replacing damaged myocardium and vascular tissues. Despite these findings, the endogenous actions of cardiovascular stem cells and cardiovascular progenitor cells appear to be insufficient to protect against cardiovascular disease in older individuals. Because recent evidence suggests that cardiovascular stem cells and cardiovascular progenitor cells are subject to age-associated changes that impair their function, these changes may contribute to the dysregulation of endogenous cardiovascular repair mechanisms in the aging heart and vasculature. Here we present the evidence for the impact of aging on cardiovascular stem cell/cardiovascular progenitor cell function and its potential importance in the increased severity of cardiovascular pathophysiology observed in the geriatric population.


Assuntos
Envelhecimento/fisiologia , Fenômenos Fisiológicos Cardiovasculares , Regeneração/fisiologia , Células-Tronco/fisiologia , Envelhecimento/patologia , Animais , Doenças Cardiovasculares/patologia , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/citologia , Sistema Cardiovascular/patologia , Senescência Celular/fisiologia , Humanos , Células-Tronco/citologia
8.
Physiol Genomics ; 26(3): 202-8, 2006 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-16705020

RESUMO

Functional proteomic strategies offer unique advantages over current molecular array approaches, as the epitopes identified can directly provide bioactive peptides for investigational and/or translational applications. The vascular endothelium is well suited to proteomic assessment by in vivo phage display, but extensive enrichment and sequencing steps limit its application for high throughput molecular profiling. To overcome these limitations we developed a quantitative PCR (Q-PCR) strategy to allow the rapid quantification of in vivo phage binding. Primers were designed for distinct clones selected from a defined phage pool to probe for age-associated changes in cardiac vascular epitopes. Sensitivity and specificity of the primer sets were tested and confirmed in vitro. Q-PCR quantification of phage in vivo confirmed the preferential homing of all phage clones to the young rather than old cardiac vasculature and demonstrated a close correlation with phage measurements previously determined using traditional bacterial-based titration methods. This Q-PCR approach provides quantification of phage within hours of phage injection and may therefore be used for rapid, high throughput analysis of binding of defined phage sequences both in vivo and in vitro, complementing nonbiased phage approaches for the proteomic mapping of vascular beds and other tissues.


Assuntos
Biblioteca de Peptídeos , Reação em Cadeia da Polimerase/métodos , Proteômica/métodos , Envelhecimento , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Vasos Coronários/metabolismo , Primers do DNA/genética , Imuno-Histoquímica , Camundongos , Dados de Sequência Molecular , Miócitos Cardíacos/metabolismo , Fragmentos de Peptídeos/análise , Fragmentos de Peptídeos/genética , Reprodutibilidade dos Testes , Homologia de Sequência de Aminoácidos
9.
FASEB J ; 20(6): 717-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16461331

RESUMO

Microenvironmental cues mediate postnatal neovascularization via modulation of endothelial cell and bone marrow-derived endothelial progenitor cell (EPC) activity. Numerous signals regulate the activity of both of these cell types in response to vascular injury, which suggests that parallel mechanisms regulate angiogenesis in the vascular beds of both the heart and bone marrow. To identify mediators of such shared pathways, in vivo bone marrow/cardiac phage display biopanning was performed and led to the identification of tenascin-C as a candidate protein. Functionally, tenascin-C inhibits cardiac endothelial cell spreading and enhances migration in response to angiogenic growth factors. Analysis of human coronary thrombi revealed tenascin-C protein expression colocalized with the endothelial cell/EPC marker Tie-2 in intrathrombi vascular channels. Immunostains in the rodent heart demonstrated that tenascin-C also colocalizes with EPCs homing to sites of cardiac angiogenic induction. To determine the importance of tenascin-C in cardiac neovascularization, we used an established cardiac transplantation model and showed that unlike wild-type mice, tenascin-C-/- mice fail to vascularize cardiac allografts. This demonstrates for the first time that tenascin-C is essential for postnatal cardiac angiogenic function. Together, our data highlight the role of tenascin-C as a microenvironmental regulator of cardiac endothelial/EPC activity.


Assuntos
Vasos Coronários/crescimento & desenvolvimento , Endotélio Vascular/fisiologia , Neovascularização Fisiológica/fisiologia , Tenascina/metabolismo , Envelhecimento , Animais , Células da Medula Óssea/metabolismo , Adesão Celular , Movimento Celular , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Knockout , Miocárdio/metabolismo , Fenótipo , Ratos , Ratos Endogâmicos F344 , Tenascina/genética , Trombose/metabolismo
10.
Physiol Genomics ; 24(3): 191-7, 2006 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-16352696

RESUMO

Aging is associated with shifts in autocrine and paracrine pathways in the cardiac vasculature that may contribute to the risk of cardiovascular disease in older persons. To elucidate the molecular basis of these changes in vivo, phage-display biopanning of 3- and 18-mo-old mouse hearts was performed that identified peptide epitopes with homology to brain-derived neurotrophic factor (BDNF) in old but not young phage pools. Quantification of cardiac phage binding by titration and immunostaining after injection with BDNF-like phage identified a twofold increased density of the BDNF receptor, truncated Trk B, in the aging hearts. Studies focused on the receptor ligand using a rat model of transient myocardial ischemia revealed increases in cardiac BDNF associated with local mononuclear infiltrates in 24- but not 4-mo-old rats. To investigate these changes, both 4- and 24-mo-old rat hearts were treated with intramyocardial injections of BDNF (or PBS control), demonstrating significant inflammatory increases with activated macrophage (ED1+) in BDNF-treated aging hearts compared with aging controls and similarly treated young hearts. Additional studies with permanent coronary occlusion following intramyocardial growth factor pretreatment revealed that BDNF significantly increased the extent of myocardial injury in older rat hearts (BDNF 35 +/- 10% vs. PBS 16.2 +/- 7.9% left ventricular injury; P < 0.05) without affecting younger hearts (BDNF 15 +/- 5.1% vs. PBS 14.5 +/- 6.0% left ventricular injury). Overall, these studies suggest that age-associated changes in BDNF-Trk B pathways may predispose the aging heart to increased injury after acute myocardial infarction and potentially contribute to the enhanced severity of cardiovascular disease in older individuals.


Assuntos
Envelhecimento/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Inflamação/metabolismo , Miocárdio/metabolismo , Animais , Feminino , Camundongos , Infarto do Miocárdio/metabolismo , Miocárdio/enzimologia , Biblioteca de Peptídeos , Ratos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor trkB/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo
11.
J Exp Med ; 199(6): 797-804, 2004 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-15007092

RESUMO

Pretreatment of rodent hearts with platelet-derived growth factor (PDGF)-AB decreases myocardial injury after coronary occlusion. However, PDGF-AB cardioprotection is diminished in older animals, suggesting that downstream elements mediating and/or synergizing the actions of PDGF-AB may be limited in aging cardiac vasculature. In vitro PDGF-AB induced vascular endothelial growth factor (VEGF) and angiopoietin (Ang)-2 expression in 4-mo-old rat cardiac endothelial cells, but not in 24-mo-old heart cells. In vivo injection of young hearts with PDGF-AB increased densities of microvessels staining for VEGF and its receptor, Flk-1, and Ang-2 and its receptor, Tie-2, as well as PDGF receptor (PDGFR)-alpha. In older hearts, PDGF-AB-mediated induction was primarily limited to PDGFR-alpha. Studies in a murine cardiac transplantation model demonstrated that synergist interactions of PDGF-AB plus VEGF plus Ang-2 (PVA) provided an immediate restoration of senescent cardiac vascular function. Moreover, PVA injection in young rat hearts, but not PDGF-AB alone or other cytokine combinations, at the time of coronary occlusion suppressed acute myocardial cell death by >50%. However, PVA also reduced the extent of myocardial infarction with an age-associated cardioprotective benefit (4-mo-old with 45% reduction vs. 24-mo-old with 24%; P < 0.05). These studies showed that synergistic cytokine pathways augmenting the actions of PDGF-AB are limited in older hearts, suggesting that strategies based on these interactions may provide age-dependent clinical cardiovascular benefit.


Assuntos
Envelhecimento/metabolismo , Angiopoietina-2/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Fator de Crescimento Derivado de Plaquetas/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/uso terapêutico , Animais , Cardiotônicos/uso terapêutico , Células Cultivadas , Doença das Coronárias/complicações , Primers do DNA , Sinergismo Farmacológico , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Ratos , Ratos Endogâmicos F344 , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor TIE-2/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Dev Dyn ; 228(2): 161-72, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14517988

RESUMO

Proper heart development requires patterning across the myocardial wall. Early myocardial patterning is characterized by a transmural subdivision of the myocardium into an outer, highly mitotic, compact zone and an inner, trabecular zone with lower mitotic activity. We have shown previously that fibroblast growth factor receptor (FGFR) -mediated signaling is central to myocyte proliferation in the developing heart. Consistent with this, FGFR-1 and FGF2 are more highly expressed in myocytes of the compact zone. However, the mechanism that regulates the transmural pattern of myocyte proliferation and expression of these mitogenic factors is unknown. The present study examined whether this transmural patterning occurs in a myocardium-autonomous manner or by signals from the epicardium. Microsurgical inhibition of epicardium formation in the embryonic chick gives rise to a decrease in myocyte proliferation, accounting for a thinner compact myocardium. We show that the transmural pattern of myocyte mitotic activity is maintained in these hearts. Consistent with this, the expression patterns of FGF1, FGF2, and FGFR-1 across the myocardium persist in the absence of the epicardium. However, FGF2 and FGFR-1 mRNA levels are reduced in proportion to the depletion of epicardium. The results suggest that epicardium-derived signals are essential for maintenance of the correct amount of myocyte proliferation in the compact myocardium, by means of levels of mitogen expression in the myocardium. However, initiation and maintenance of transmural patterning of the myocardium occurs largely independently of the epicardium.


Assuntos
Fator 2 de Crescimento de Fibroblastos/metabolismo , Coração/embriologia , Miócitos Cardíacos/metabolismo , Pericárdio/fisiologia , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Apoptose , Divisão Celular , Embrião de Galinha , Fator 1 de Crescimento de Fibroblastos/metabolismo , Imuno-Histoquímica , Ligantes , Microcirurgia , Mitógenos/metabolismo , Miócitos Cardíacos/citologia , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
13.
Dev Biol ; 251(1): 167-77, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12413906

RESUMO

Cardiac neural crest cells are essential for normal development of the great vessels and the heart, giving rise to a range of cell types, including both neuronal and non-neuronal adventitial cells and smooth muscle. Endothelin (ET) signaling plays an important role in the development of cardiac neural crest cell lineages, yet the underlying mechanisms that act to control their migration, differentiation, and proliferation remain largely unclear. We examined the expression patterns of the receptor, ET(A), and the ET-specific converting enzyme, ECE-1, in the pharyngeal arches and great vessels of the developing chick embryo. In situ hybridization analysis revealed that, while ET(A) is expressed in the pharyngeal arch mesenchyme, populated by cardiac neural crest cells, ECE-1 expression is localized to the outermost ectodermal cells of the arches and then to the innermost endothelial cells of the great vessels. This dynamic pattern of expression suggests that only a subpopulation of neural crest cells in these regions is responsive to ET signaling at particular developmental time points. To test this, retroviral gene delivery was used to constitutively express preproET-1, a precursor of mature ET-1 ligand, in the cardiac neural crest. This resulted in a selective expansion of the outermost, adventitial cell population in the great vessels. In contrast, neither differentiation nor proliferation of neural crest-derived smooth muscle cells was significantly affected. These results suggest that constitutive expression of exogenous preproET-1 in the cardiac neural crest results in expansion restricted to an adventitial cell population of the developing great vessels.


Assuntos
Sistema Cardiovascular/embriologia , Endotelinas/genética , Coração/embriologia , Crista Neural/embriologia , Precursores de Proteínas/genética , Animais , Região Branquial/embriologia , Sistema Cardiovascular/citologia , Contagem de Células , Diferenciação Celular/genética , Embrião de Galinha , Endotelina-1 , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...